Skip to main content
Top
Published in: BMC Cancer 1/2016

Open Access 01-12-2016 | Research article

Epigenetic silencing of miR-181c by DNA methylation in glioblastoma cell lines

Authors: Erandi Ayala-Ortega, Rodrigo Arzate-Mejía, Rosario Pérez-Molina, Edgar González-Buendía, Karin Meier, Georgina Guerrero, Félix Recillas-Targa

Published in: BMC Cancer | Issue 1/2016

Login to get access

Abstract

Background

Post-transcriptional regulation by microRNAs is recognized as one of the major pathways for the control of cellular homeostasis. Less well understood is the transcriptional and epigenetic regulation of genes encoding microRNAs. In the present study we addressed the epigenetic regulation of the miR-181c in normal and malignant brain cells.

Methods

To explore the epigenetic regulation of the miR-181c we evaluated its expression using RT-qPCR and the in vivo binding of the CCCTC-binding factor (CTCF) to its regulatory region in different glioblastoma cell lines. DNA methylation survey, chromatin immunoprecipitation and RNA interference assays were used to assess the role of CTCF in the miR-181c epigenetic silencing.

Results

We found that miR-181c is downregulated in glioblastoma cell lines, as compared to normal brain tissues. Loss of expression correlated with a notorious gain of DNA methylation at the miR-181c promoter region and the dissociation of the multifunctional nuclear factor CTCF. Taking advantage of the genomic distribution of CTCF in different cell types we propose that CTCF has a local and cell type specific regulatory role over the miR-181c and not an architectural one through chromatin loop formation. This is supported by the depletion of CTCF in glioblastoma cells affecting the expression levels of NOTCH2 as a target of miR-181c.

Conclusion

Together, our results point to the epigenetic role of CTCF in the regulation of microRNAs implicated in tumorigenesis.
Appendix
Available only for authorised users
Literature
3.
go back to reference Benetatos L, Voulgaris E, Vartholomatos G, Hatzimichael E. Non-coding RNAs and EZH2 interactions in cancer: long and short tales from the transcriptome. Int J Cancer. 2013;133(2):267–74.CrossRefPubMed Benetatos L, Voulgaris E, Vartholomatos G, Hatzimichael E. Non-coding RNAs and EZH2 interactions in cancer: long and short tales from the transcriptome. Int J Cancer. 2013;133(2):267–74.CrossRefPubMed
4.
go back to reference Di Leva G, Garofalo M, Croce CM. MicroRNAs in cancer. Ann Rev Pathol Mech Dis. 2014;9:287–314.CrossRef Di Leva G, Garofalo M, Croce CM. MicroRNAs in cancer. Ann Rev Pathol Mech Dis. 2014;9:287–314.CrossRef
6.
go back to reference Fabbri M, Garzon R, Cimmino A, Liu Z, Zanesi N, Callegari E, et al. MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferase 3A and 3B. Proc Natl Acad Sci U S A. 2007;104(40):15805–10.CrossRefPubMedPubMedCentral Fabbri M, Garzon R, Cimmino A, Liu Z, Zanesi N, Callegari E, et al. MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferase 3A and 3B. Proc Natl Acad Sci U S A. 2007;104(40):15805–10.CrossRefPubMedPubMedCentral
7.
go back to reference Garzon R, Liu S, Fabbri M, Liu Z, Heaphy CE, Callegari E, et al. MicroRNA-29b induces global DNA hypomethylation and tumor suppressor gene reexpression in acute myeloid leukemia by targeting directly DNMT3A and 3B and indirectly DNMT1. Blood. 2009;113(25):6411–8.CrossRefPubMedPubMedCentral Garzon R, Liu S, Fabbri M, Liu Z, Heaphy CE, Callegari E, et al. MicroRNA-29b induces global DNA hypomethylation and tumor suppressor gene reexpression in acute myeloid leukemia by targeting directly DNMT3A and 3B and indirectly DNMT1. Blood. 2009;113(25):6411–8.CrossRefPubMedPubMedCentral
8.
go back to reference Zhao B, Bian EB, Li J, Li J. New advances of microRNAs in glioma stem cells, with special emphasis on aberrant methylation of microRNAs. J Cell Physiol. 2014;229(9):1141–7.CrossRefPubMed Zhao B, Bian EB, Li J, Li J. New advances of microRNAs in glioma stem cells, with special emphasis on aberrant methylation of microRNAs. J Cell Physiol. 2014;229(9):1141–7.CrossRefPubMed
9.
go back to reference Asuthkar S, Velpula KK, Chetty C, Gorantla B, Rao JS. Epigenetic regulation of miRNA-211 by MMP-9 governs glioma cell apoptosis, chemosensitivity and radiosensitivity. Oncotarget. 2012;3(11):1439–54.CrossRefPubMedPubMedCentral Asuthkar S, Velpula KK, Chetty C, Gorantla B, Rao JS. Epigenetic regulation of miRNA-211 by MMP-9 governs glioma cell apoptosis, chemosensitivity and radiosensitivity. Oncotarget. 2012;3(11):1439–54.CrossRefPubMedPubMedCentral
10.
go back to reference Ying Z, Li Y, Wu J, Zhu X, Yang Y, Tian H, et al. Loss of miR-204 expression enhances glioma migration and stem cell-like phenotype. Cancer Res. 2013;73(2):990–9.CrossRefPubMedPubMedCentral Ying Z, Li Y, Wu J, Zhu X, Yang Y, Tian H, et al. Loss of miR-204 expression enhances glioma migration and stem cell-like phenotype. Cancer Res. 2013;73(2):990–9.CrossRefPubMedPubMedCentral
11.
go back to reference Lee HK, Bier A, Cazacu S, Finniss S, Xiang C, Twito H, et al. MicroRNA-145 is downregulated in glial tumors and regulates glioma cell migration by targeting connective tissue growth factor. PLoS ONE. 2013;8(2), e54652.CrossRefPubMedPubMedCentral Lee HK, Bier A, Cazacu S, Finniss S, Xiang C, Twito H, et al. MicroRNA-145 is downregulated in glial tumors and regulates glioma cell migration by targeting connective tissue growth factor. PLoS ONE. 2013;8(2), e54652.CrossRefPubMedPubMedCentral
12.
go back to reference Bier A, Giladi N, Kronfeld N, Lee HK, Cazacu S, Finniss C, et al. MicroRNA-137 is downregulated in glioblastoma and inhibits the stemness of glioma stem cells by targeting RTVP-1. Oncotarget. 2013;4(5):665–76.CrossRefPubMedPubMedCentral Bier A, Giladi N, Kronfeld N, Lee HK, Cazacu S, Finniss C, et al. MicroRNA-137 is downregulated in glioblastoma and inhibits the stemness of glioma stem cells by targeting RTVP-1. Oncotarget. 2013;4(5):665–76.CrossRefPubMedPubMedCentral
13.
go back to reference Lee HK, Finniss S, Cazacu S, Bucris E, Ziv-Av A, Xiang C, et al. Mesenchymal stem cells deliver synthetic microRNA mimics to glioma cells and glioma stem cells and inhibit their cell migration and self-renewal. Oncotarget. 2013;4(2):346–61.CrossRefPubMedPubMedCentral Lee HK, Finniss S, Cazacu S, Bucris E, Ziv-Av A, Xiang C, et al. Mesenchymal stem cells deliver synthetic microRNA mimics to glioma cells and glioma stem cells and inhibit their cell migration and self-renewal. Oncotarget. 2013;4(2):346–61.CrossRefPubMedPubMedCentral
14.
go back to reference Filippova GN, Fagerlie S, Klenova EM, Myers C, Dehner Y, Goodwin G, et al. An exceptionally conserved transcriptional repressor, CTCF, employs different combinations of zinc fingers to bind diverged promoter sequences of avian and mammalian c-myc oncogenes. Mol Cell Biol. 1996;16(6):2802–13.CrossRefPubMedPubMedCentral Filippova GN, Fagerlie S, Klenova EM, Myers C, Dehner Y, Goodwin G, et al. An exceptionally conserved transcriptional repressor, CTCF, employs different combinations of zinc fingers to bind diverged promoter sequences of avian and mammalian c-myc oncogenes. Mol Cell Biol. 1996;16(6):2802–13.CrossRefPubMedPubMedCentral
15.
go back to reference Klenova EM, Nicolas RH, Paterson HF, Carne AF, Heath CM, Goodwin GH, et al. CTCF, a conserved nuclear factor required for optimal transcriptional activity of the chicken c-myc gene, is an 11-Zn-finger protein differentially expressed in multiple forms. Mol Cell Biol. 1993;13(12):7612–24.CrossRefPubMedPubMedCentral Klenova EM, Nicolas RH, Paterson HF, Carne AF, Heath CM, Goodwin GH, et al. CTCF, a conserved nuclear factor required for optimal transcriptional activity of the chicken c-myc gene, is an 11-Zn-finger protein differentially expressed in multiple forms. Mol Cell Biol. 1993;13(12):7612–24.CrossRefPubMedPubMedCentral
16.
go back to reference MacPherson MJ, Beatty LG, Zhou W, Du M, Sadowski PD. The CTCF insulator protein is posttranslationally modified by SUMO. Mol Cell Biol. 2009;29(3):714–25.CrossRefPubMedPubMedCentral MacPherson MJ, Beatty LG, Zhou W, Du M, Sadowski PD. The CTCF insulator protein is posttranslationally modified by SUMO. Mol Cell Biol. 2009;29(3):714–25.CrossRefPubMedPubMedCentral
17.
go back to reference Yu W, Ginjala V, Pant V, Chernukhin I, Whitehead J, Docquier F, et al. Poly(ADP-ribosyl)ation regulates CTCF-dependent chromatin insulation. Nat Genet. 2004;36(10):1105–10.CrossRefPubMed Yu W, Ginjala V, Pant V, Chernukhin I, Whitehead J, Docquier F, et al. Poly(ADP-ribosyl)ation regulates CTCF-dependent chromatin insulation. Nat Genet. 2004;36(10):1105–10.CrossRefPubMed
19.
go back to reference Dixon JR, Selvaraj S, Yue F, Kim A, Li Y, Shen Y, et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature. 2012;485(7398):376–80.CrossRefPubMedPubMedCentral Dixon JR, Selvaraj S, Yue F, Kim A, Li Y, Shen Y, et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature. 2012;485(7398):376–80.CrossRefPubMedPubMedCentral
20.
go back to reference Zuin J, Dixon JR, van der Reijden MI, Ye Z, Kolovos P, Brouwer RW, et al. Cohesin and CTCF differentially affect chromatin architecture and gene expression in human cells. Proc Natl Acad Sci U S A. 2014;111(3):996–1001.CrossRefPubMedPubMedCentral Zuin J, Dixon JR, van der Reijden MI, Ye Z, Kolovos P, Brouwer RW, et al. Cohesin and CTCF differentially affect chromatin architecture and gene expression in human cells. Proc Natl Acad Sci U S A. 2014;111(3):996–1001.CrossRefPubMedPubMedCentral
21.
go back to reference Zlatanova J, Caiafa P. CTCF and its protein partners: divide and rule? J Cell Sci. 2009;122(Pt 9):1275–84.CrossRefPubMed Zlatanova J, Caiafa P. CTCF and its protein partners: divide and rule? J Cell Sci. 2009;122(Pt 9):1275–84.CrossRefPubMed
22.
go back to reference Choi NM, Feeney AJ. CTCF and ncRNA regulate the three-dimensional structure of antigen receptor loci to facilitate V(D)J recombination. Front Immunol. 2014;5:49.CrossRefPubMedPubMedCentral Choi NM, Feeney AJ. CTCF and ncRNA regulate the three-dimensional structure of antigen receptor loci to facilitate V(D)J recombination. Front Immunol. 2014;5:49.CrossRefPubMedPubMedCentral
23.
go back to reference Saldaña-Meyer R, González-Buendía E, Guerrero G, Narendra V, Bonasio R, Recillas-Targa F, et al. CTCF regulates the human p53 gene through direct interaction with its natural antisense transcript, Wrap53. Genes Dev. 2014;28(7):723–34.CrossRefPubMedPubMedCentral Saldaña-Meyer R, González-Buendía E, Guerrero G, Narendra V, Bonasio R, Recillas-Targa F, et al. CTCF regulates the human p53 gene through direct interaction with its natural antisense transcript, Wrap53. Genes Dev. 2014;28(7):723–34.CrossRefPubMedPubMedCentral
24.
go back to reference Recillas-Targa F, De La Rosa-Velázquez IA, Soto-Reyes R, Benítez-Bribiesca L. Epigenetic boundaries of tumour suppressor gene promoters: the CTCF connection and its role in carcinogenesis. J Cell Mol Med. 2006;10(3):554–68.CrossRefPubMedPubMedCentral Recillas-Targa F, De La Rosa-Velázquez IA, Soto-Reyes R, Benítez-Bribiesca L. Epigenetic boundaries of tumour suppressor gene promoters: the CTCF connection and its role in carcinogenesis. J Cell Mol Med. 2006;10(3):554–68.CrossRefPubMedPubMedCentral
25.
go back to reference Recillas-Targa F, De La Rosa-Velázquez IA, Soto-Reyes E. Insulation of tumor suppressor genes by the nuclear factor CTCF. Biochem Cell Biol. 2011;89(5):479–88.CrossRefPubMed Recillas-Targa F, De La Rosa-Velázquez IA, Soto-Reyes E. Insulation of tumor suppressor genes by the nuclear factor CTCF. Biochem Cell Biol. 2011;89(5):479–88.CrossRefPubMed
26.
go back to reference Engel N, West AG, Felsenfeld G, Bartolomei MS. Antagonism between DNA hypermethylation and enhancer-blocking activity at the H19 DMD is uncovered by CpG mutations. Nat Genet. 2004;36(8):883–8.CrossRefPubMed Engel N, West AG, Felsenfeld G, Bartolomei MS. Antagonism between DNA hypermethylation and enhancer-blocking activity at the H19 DMD is uncovered by CpG mutations. Nat Genet. 2004;36(8):883–8.CrossRefPubMed
27.
go back to reference Chang J, Zhang B, Heatch H, Galjart N, Wang X, Milbrandt J. Nicotinamide adenine dinucleotide (NAD)-regulated DNA methylation alters CCCTC-binding factor (CTCF)/cohesin binding and transcription at the BDNF locus. Proc Natl Acad Sci U S A. 2010;107(50):21836–41.CrossRefPubMedPubMedCentral Chang J, Zhang B, Heatch H, Galjart N, Wang X, Milbrandt J. Nicotinamide adenine dinucleotide (NAD)-regulated DNA methylation alters CCCTC-binding factor (CTCF)/cohesin binding and transcription at the BDNF locus. Proc Natl Acad Sci U S A. 2010;107(50):21836–41.CrossRefPubMedPubMedCentral
28.
go back to reference Wang H, Maurano MT, Qu H, Varley KE, Gertz J, Pauli F, et al. Widespread plasticity in CTCF occupancy linked to DNA methylation. Genome Res. 2012;22(9):1680–8.CrossRefPubMedPubMedCentral Wang H, Maurano MT, Qu H, Varley KE, Gertz J, Pauli F, et al. Widespread plasticity in CTCF occupancy linked to DNA methylation. Genome Res. 2012;22(9):1680–8.CrossRefPubMedPubMedCentral
30.
go back to reference Soto-Reyes E, Recillas-Targa F. Epigenetic regulation of the human p53 gene promoter by the CTCF transcription factor in transformed cell lines. Oncogene. 2010;29(15):2217–27.CrossRefPubMed Soto-Reyes E, Recillas-Targa F. Epigenetic regulation of the human p53 gene promoter by the CTCF transcription factor in transformed cell lines. Oncogene. 2010;29(15):2217–27.CrossRefPubMed
31.
go back to reference de Souza Rocha Simonini P, Breiling A, Gupta N, Malekpour M, Youns M, Omranipour R, et al. Epigenetically deregulated microRNA-375 is involved in a positive feedback loop with estrogen receptor alpha in breast cancer cells. Cancer Res. 2010;70(22):9175–84.CrossRefPubMed de Souza Rocha Simonini P, Breiling A, Gupta N, Malekpour M, Youns M, Omranipour R, et al. Epigenetically deregulated microRNA-375 is involved in a positive feedback loop with estrogen receptor alpha in breast cancer cells. Cancer Res. 2010;70(22):9175–84.CrossRefPubMed
32.
go back to reference Tata PR, Tata NR, Kühl M, Sirbu IO. Identification of a novel epigenetic regulatory region within the pluripotency associated microRNA cluster, EEmiRC. Nucleic Acids Res. 2011;39(9):3574–81.CrossRefPubMedPubMedCentral Tata PR, Tata NR, Kühl M, Sirbu IO. Identification of a novel epigenetic regulatory region within the pluripotency associated microRNA cluster, EEmiRC. Nucleic Acids Res. 2011;39(9):3574–81.CrossRefPubMedPubMedCentral
33.
go back to reference Lakomy R, Sana J, Hankeova S, Fadrus P, Kren L, Lzicarova E, et al. MiR-195, miR-196b, miR-181c, miR-21 expression levels and O-6-methylguanine-DNA methyltransferase methylation status are associated with clinical outcome in glioblastoma patients. Cancer Sci. 2011;102(12):2186–90.CrossRefPubMed Lakomy R, Sana J, Hankeova S, Fadrus P, Kren L, Lzicarova E, et al. MiR-195, miR-196b, miR-181c, miR-21 expression levels and O-6-methylguanine-DNA methyltransferase methylation status are associated with clinical outcome in glioblastoma patients. Cancer Sci. 2011;102(12):2186–90.CrossRefPubMed
34.
go back to reference Ruan J, Lou S, Dai Q, Mao D, Ji J, Sun X. Tumor suppressor miR-181c attenuates proliferation, invasion and self-renewal abilities in glioblastoma. NeuroReport. 2015;26(2):66–73.CrossRefPubMed Ruan J, Lou S, Dai Q, Mao D, Ji J, Sun X. Tumor suppressor miR-181c attenuates proliferation, invasion and self-renewal abilities in glioblastoma. NeuroReport. 2015;26(2):66–73.CrossRefPubMed
35.
go back to reference Sand M, Skrygan M, Sand D, Georgas D, Hahn SA, Gambichler T, et al. Expression of microRNAs in basal cell carcinoma. Br J Dermatol. 2012;167(4):847–55.CrossRefPubMed Sand M, Skrygan M, Sand D, Georgas D, Hahn SA, Gambichler T, et al. Expression of microRNAs in basal cell carcinoma. Br J Dermatol. 2012;167(4):847–55.CrossRefPubMed
36.
go back to reference Jones KB, Salah Z, Del Mare S, Galasso M, Gaudio E, Nuovo GJ, et al. miRNA signature associate with pathogenesis and progression of osteosarcoma. Cancer Res. 2012;72(7):1865–77.CrossRefPubMedPubMedCentral Jones KB, Salah Z, Del Mare S, Galasso M, Gaudio E, Nuovo GJ, et al. miRNA signature associate with pathogenesis and progression of osteosarcoma. Cancer Res. 2012;72(7):1865–77.CrossRefPubMedPubMedCentral
37.
go back to reference Cui MH, Hou XL, Lei XY, Mu FH, Yang GB, Yue L, et al. Upregulation of microRNA 181c expression in gastric cancer tissues and plasma. Asian Pac J Cancer Prev. 2013;14(5):3063–6.CrossRefPubMed Cui MH, Hou XL, Lei XY, Mu FH, Yang GB, Yue L, et al. Upregulation of microRNA 181c expression in gastric cancer tissues and plasma. Asian Pac J Cancer Prev. 2013;14(5):3063–6.CrossRefPubMed
38.
go back to reference Dávalos-Salas M, Furlan-Magaril M, González-Buendía E, Valdes-Quezada C, Ayala-Ortega E, Recillas-Targa F. Gain of DNA methylation is enhanced in the absence of CTCF at the human retinoblastoma gene promoter. BMC Cancer. 2011;11:232.CrossRefPubMedPubMedCentral Dávalos-Salas M, Furlan-Magaril M, González-Buendía E, Valdes-Quezada C, Ayala-Ortega E, Recillas-Targa F. Gain of DNA methylation is enhanced in the absence of CTCF at the human retinoblastoma gene promoter. BMC Cancer. 2011;11:232.CrossRefPubMedPubMedCentral
39.
40.
go back to reference Ozsolak F, Poling LL, Wang Z, Liu H, Liu XS, Roeder RG, et al. Chromatin structure analyses identify miRNA promoters. Genes Dev. 2008;22(22):3172–83.CrossRefPubMedPubMedCentral Ozsolak F, Poling LL, Wang Z, Liu H, Liu XS, Roeder RG, et al. Chromatin structure analyses identify miRNA promoters. Genes Dev. 2008;22(22):3172–83.CrossRefPubMedPubMedCentral
41.
go back to reference Cui H, Niemitz EL, Ravenel JD, Onyango P, Brandenburg SA, Lobanenkov VV, et al. Loss of imprinting of insulin-like growth factor-II in Wilms’ tumor commonly involves altered methylation but not mutations of CTCF or its binding site. Cancer Res. 2001;61(13):4947–50.PubMed Cui H, Niemitz EL, Ravenel JD, Onyango P, Brandenburg SA, Lobanenkov VV, et al. Loss of imprinting of insulin-like growth factor-II in Wilms’ tumor commonly involves altered methylation but not mutations of CTCF or its binding site. Cancer Res. 2001;61(13):4947–50.PubMed
42.
go back to reference Sexton T, Cavalli G. The role of chromatin domains in shaping the functional genome. Cell. 2015;160(6):1049–59.CrossRefPubMed Sexton T, Cavalli G. The role of chromatin domains in shaping the functional genome. Cell. 2015;160(6):1049–59.CrossRefPubMed
43.
go back to reference Rao SS, Huntley MH, Durand NC, Stamenova EK, Bochkov ID, Robinsin JT, et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell. 2014;159(7):1665–80.CrossRefPubMed Rao SS, Huntley MH, Durand NC, Stamenova EK, Bochkov ID, Robinsin JT, et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell. 2014;159(7):1665–80.CrossRefPubMed
44.
45.
go back to reference Recillas-Targa F. Interdependency between genetic and epigenetic regulatory defects in cancer. Methods Mol Biol. 2014;1165:33–52.CrossRefPubMed Recillas-Targa F. Interdependency between genetic and epigenetic regulatory defects in cancer. Methods Mol Biol. 2014;1165:33–52.CrossRefPubMed
46.
go back to reference Au SL, Wong CC, Lee JM, Fan DN, Tsang FH, Ng IO, et al. Enhancer of zeste homolog 2 epigenetically silences multiple tumor suppressor microRNAs to promote liver cancer metastasis. Hepatalogy. 2012;56(2):622–31.CrossRef Au SL, Wong CC, Lee JM, Fan DN, Tsang FH, Ng IO, et al. Enhancer of zeste homolog 2 epigenetically silences multiple tumor suppressor microRNAs to promote liver cancer metastasis. Hepatalogy. 2012;56(2):622–31.CrossRef
47.
go back to reference Cao Q, Manu RS, Russo N, Scanlon CS, Tsodikov A, Jing X, et al. Coordinated regulation of polycomb group complexes through microRNAs in cancer. Cancer Cell. 2011;20(2):187–99.CrossRefPubMedPubMedCentral Cao Q, Manu RS, Russo N, Scanlon CS, Tsodikov A, Jing X, et al. Coordinated regulation of polycomb group complexes through microRNAs in cancer. Cancer Cell. 2011;20(2):187–99.CrossRefPubMedPubMedCentral
48.
go back to reference Tiffen JC, Bailey CG, Marshall AD, Metierre C, Feng Y, Wang Q, et al. The cancer-testis antigen BORIS phenocopies the tumor suppressor CTCF in normal and neoplastic cells. Int J Cancer. 2013;133(7):1603–14.CrossRefPubMed Tiffen JC, Bailey CG, Marshall AD, Metierre C, Feng Y, Wang Q, et al. The cancer-testis antigen BORIS phenocopies the tumor suppressor CTCF in normal and neoplastic cells. Int J Cancer. 2013;133(7):1603–14.CrossRefPubMed
49.
go back to reference Nakahashi H, Kwon KR, Resch W, Vian L, Dose M, Stavreva D, et al. A genome-map of CTCF multivalency redefines the CTCF code. Cell Rep. 2013;3(5):1678–89.CrossRefPubMedPubMedCentral Nakahashi H, Kwon KR, Resch W, Vian L, Dose M, Stavreva D, et al. A genome-map of CTCF multivalency redefines the CTCF code. Cell Rep. 2013;3(5):1678–89.CrossRefPubMedPubMedCentral
50.
go back to reference Kemp CJ, Moore JM, Moser R, Bernard B, Teater M, Smith LE, et al. CTCF haploinsufficiency destabilizes DNA methylation and predisposes to cancer. Cell Rep. 2014;7(4):1020–9.CrossRefPubMedPubMedCentral Kemp CJ, Moore JM, Moser R, Bernard B, Teater M, Smith LE, et al. CTCF haploinsufficiency destabilizes DNA methylation and predisposes to cancer. Cell Rep. 2014;7(4):1020–9.CrossRefPubMedPubMedCentral
51.
go back to reference Lawrence MS, Stojanov P, Mermel CH, Robinson JT, Garraway LA, Golub TR, et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature. 2014;505(7484):495–501.CrossRefPubMedPubMedCentral Lawrence MS, Stojanov P, Mermel CH, Robinson JT, Garraway LA, Golub TR, et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature. 2014;505(7484):495–501.CrossRefPubMedPubMedCentral
Metadata
Title
Epigenetic silencing of miR-181c by DNA methylation in glioblastoma cell lines
Authors
Erandi Ayala-Ortega
Rodrigo Arzate-Mejía
Rosario Pérez-Molina
Edgar González-Buendía
Karin Meier
Georgina Guerrero
Félix Recillas-Targa
Publication date
01-12-2016
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2016
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-016-2273-6

Other articles of this Issue 1/2016

BMC Cancer 1/2016 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine