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

Open Access 01-12-2016 | Research article

Involvement of DNMT 3B promotes epithelial-mesenchymal transition and gene expression profile of invasive head and neck squamous cell carcinomas cell lines

Authors: Li-Hsuen Chen, Wen-Lin Hsu, Yen-Ju Tseng, Dai-Wei Liu, Ching-Feng Weng

Published in: BMC Cancer | Issue 1/2016

Login to get access

Abstract

Background

The 5-year overall survival rates for head and neck cancer (HNC) relies on distant metastasis. Importantly, the epithelial-mesenchymal transition (EMT) is believed to be an initial step of metastasis. However, the relationship of epigenetic with EMT formation is still unexplored in HNC. This study focuses on invasive subclones of HNC cell lines through the simulation of invasion in vitro; and underlying mechanisms were analyzed including DNA methylation and gene expression profile.

Methods

Invasive subclones of NHC cell lines were successfully obtained using transwell coated with Matrixgel. Cells invaded through 8 μm pore several times were subcultured and examined with EMT features including morphology, EMT marker genes expression, and invasive ability. Moreover, compared the profile of genes expression in parental and invasive cells was analyzed using mRNA expression array.

Results

DNA methyltransferase 3B (DNMT 3B) was upregulated in invasive subclones and might control the 5′ region of E-cadherin (E-cad) methylation and further inhibited E-cad protein expression. Interference of DNMT 3B by siRNA or miRNA 29b could reduce EMT and cell invasion. Expression array analysis revealed the most possible involved pathways in cell invasion including arginine and proline metabolism, TGF-beta, and focal adhesion.

Conclusions

DNMT 3B might control EMT by DNA methylation manner in invasive HNC cell lines. Moreover, miR-29b mimic downregulated DNMT 3B and inhibited EMT and cell invasion indicated the role of therapeutic agent for invasive HNC. Genes identified from array data and new molecules are involved in metastasis of HNC need further validation.
Appendix
Available only for authorised users
Literature
1.
go back to reference Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61(2):69–90.CrossRefPubMed Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61(2):69–90.CrossRefPubMed
2.
go back to reference Freedman N, Schatzkin A, Leitzmann M, Hollenbeck A, Abnet C. Alcohol and head and neck cancer risk in a prospective study. Br J Cancer. 2007;96(9):1469–74.PubMedPubMedCentral Freedman N, Schatzkin A, Leitzmann M, Hollenbeck A, Abnet C. Alcohol and head and neck cancer risk in a prospective study. Br J Cancer. 2007;96(9):1469–74.PubMedPubMedCentral
3.
go back to reference Gandini S, Botteri E, Iodice S, Boniol M, Lowenfels AB, Maisonneuve P, Boyle P. Tobacco smoking and cancer: A meta‐analysis. Int J Cancer. 2008;122(1):155–64.CrossRefPubMed Gandini S, Botteri E, Iodice S, Boniol M, Lowenfels AB, Maisonneuve P, Boyle P. Tobacco smoking and cancer: A meta‐analysis. Int J Cancer. 2008;122(1):155–64.CrossRefPubMed
4.
go back to reference Hashibe M, Brennan P, Benhamou S, Castellsague X, Chen C, Curado MP, Dal Maso L, Daudt AW, Fabianova E, Wünsch-Filho V. Alcohol drinking in never users of tobacco, cigarette smoking in never drinkers, and the risk of head and neck cancer: pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. J Natl Cancer Inst. 2007;99(10):777–89.CrossRefPubMed Hashibe M, Brennan P, Benhamou S, Castellsague X, Chen C, Curado MP, Dal Maso L, Daudt AW, Fabianova E, Wünsch-Filho V. Alcohol drinking in never users of tobacco, cigarette smoking in never drinkers, and the risk of head and neck cancer: pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. J Natl Cancer Inst. 2007;99(10):777–89.CrossRefPubMed
5.
go back to reference Humans IWGotEoCRt, Organization WH, Cancer IAfRo. Betel-quid and areca-nut chewing and some areca-nut-derived nitrosamines. IARC Monogr Eval Carcinog Risks Hum. 2004;85:1–334. Humans IWGotEoCRt, Organization WH, Cancer IAfRo. Betel-quid and areca-nut chewing and some areca-nut-derived nitrosamines. IARC Monogr Eval Carcinog Risks Hum. 2004;85:1–334.
6.
go back to reference Herrero R, Castellsagué X, Pawlita M, Lissowska J, Kee F, Balaram P, Rajkumar T, Sridhar H, Rose B, Pintos J. Human papillomavirus and oral cancer: the International Agency for Research on Cancer multicenter study. J Natl Cancer Inst. 2003;95(23):1772–83.CrossRefPubMed Herrero R, Castellsagué X, Pawlita M, Lissowska J, Kee F, Balaram P, Rajkumar T, Sridhar H, Rose B, Pintos J. Human papillomavirus and oral cancer: the International Agency for Research on Cancer multicenter study. J Natl Cancer Inst. 2003;95(23):1772–83.CrossRefPubMed
7.
go back to reference Leemans CR, Tiwari R, Nauta J, Van der Waal I, Snow GB. Regional lymph node involvement and its significance in the development of distant metastases in head and neck carcinoma. Cancer. 1993;71(2):452–6.CrossRefPubMed Leemans CR, Tiwari R, Nauta J, Van der Waal I, Snow GB. Regional lymph node involvement and its significance in the development of distant metastases in head and neck carcinoma. Cancer. 1993;71(2):452–6.CrossRefPubMed
8.
go back to reference Leemans CR, Braakhuis BJ, Brakenhoff RH. The molecular biology of head and neck cancer. Nat Rev Cancer. 2011;11(1):9–22.CrossRefPubMed Leemans CR, Braakhuis BJ, Brakenhoff RH. The molecular biology of head and neck cancer. Nat Rev Cancer. 2011;11(1):9–22.CrossRefPubMed
9.
go back to reference Chambers AF, Groom AC, MacDonald IC. Metastasis: dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2002;2(8):563–72.CrossRefPubMed Chambers AF, Groom AC, MacDonald IC. Metastasis: dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2002;2(8):563–72.CrossRefPubMed
10.
go back to reference Geiger TR, Peeper DS. Metastasis mechanisms. Biochim Biophys Acta. 2009;1796(2):293–308.PubMed Geiger TR, Peeper DS. Metastasis mechanisms. Biochim Biophys Acta. 2009;1796(2):293–308.PubMed
11.
go back to reference Thiery JP. Epithelial–mesenchymal transitions in development and pathologies. Curr Opin Cell Biol. 2003;15(6):740–6.CrossRefPubMed Thiery JP. Epithelial–mesenchymal transitions in development and pathologies. Curr Opin Cell Biol. 2003;15(6):740–6.CrossRefPubMed
13.
go back to reference Thiery JP. Epithelial–mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002;2(6):442–54.CrossRefPubMed Thiery JP. Epithelial–mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002;2(6):442–54.CrossRefPubMed
14.
go back to reference Tse JC, Kalluri R. Mechanisms of metastasis: epithelial‐to‐mesenchymal transition and contribution of tumor microenvironment. J Cell Biochem. 2007;101(4):816–29.CrossRefPubMed Tse JC, Kalluri R. Mechanisms of metastasis: epithelial‐to‐mesenchymal transition and contribution of tumor microenvironment. J Cell Biochem. 2007;101(4):816–29.CrossRefPubMed
16.
go back to reference Yilmaz M, Christofori G. EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 2009;28(1–2):15–33.CrossRefPubMed Yilmaz M, Christofori G. EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 2009;28(1–2):15–33.CrossRefPubMed
17.
go back to reference Junghans D, Haas IG, Kemler R. Mammalian cadherins and protocadherins: about cell death, synapses and processing. Curr Opin Cell Biol. 2005;17(5):446–52.CrossRefPubMed Junghans D, Haas IG, Kemler R. Mammalian cadherins and protocadherins: about cell death, synapses and processing. Curr Opin Cell Biol. 2005;17(5):446–52.CrossRefPubMed
18.
go back to reference Jechlinger M, Grunert S, Tamir IH, Janda E, Lüdemann S, Waerner T, Seither P, Weith A, Beug H, Kraut N. Expression profiling of epithelial plasticity in tumor progression. Oncogene. 2003;22(46):7155–69.CrossRefPubMed Jechlinger M, Grunert S, Tamir IH, Janda E, Lüdemann S, Waerner T, Seither P, Weith A, Beug H, Kraut N. Expression profiling of epithelial plasticity in tumor progression. Oncogene. 2003;22(46):7155–69.CrossRefPubMed
19.
go back to reference Lingen M, Pinto A, Mendes R, Franchini R, Czerninski R, Tilakaratne W, Partridge M, Peterson D, Woo SB. Genetics/epigenetics of oral premalignancy: current status and future research*. Oral Dis. 2011;17(s1):7–22.CrossRefPubMed Lingen M, Pinto A, Mendes R, Franchini R, Czerninski R, Tilakaratne W, Partridge M, Peterson D, Woo SB. Genetics/epigenetics of oral premalignancy: current status and future research*. Oral Dis. 2011;17(s1):7–22.CrossRefPubMed
20.
go back to reference Luczak MW, Jagodzinski PP. The role of DNA methylation in cancer development. Folia Histochem Cytobiol. 2006;44(3):143–54.PubMed Luczak MW, Jagodzinski PP. The role of DNA methylation in cancer development. Folia Histochem Cytobiol. 2006;44(3):143–54.PubMed
21.
22.
go back to reference Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 1999;99(3):247–57.CrossRefPubMed Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 1999;99(3):247–57.CrossRefPubMed
23.
go back to reference Chang HW, Chow V, Lam KY, Wei WI, Wing Yuen AP. Loss of E‐cadherin expression resulting from promoter hypermethylation in oral tongue carcinoma and its prognostic significance. Cancer. 2002;94(2):386–92.CrossRefPubMed Chang HW, Chow V, Lam KY, Wei WI, Wing Yuen AP. Loss of E‐cadherin expression resulting from promoter hypermethylation in oral tongue carcinoma and its prognostic significance. Cancer. 2002;94(2):386–92.CrossRefPubMed
24.
go back to reference Ha PK, Califano JA. Promoter methylation and inactivation of tumour-suppressor genes in oral squamous-cell carcinoma. Lancet Oncol. 2006;7(1):77–82.CrossRefPubMed Ha PK, Califano JA. Promoter methylation and inactivation of tumour-suppressor genes in oral squamous-cell carcinoma. Lancet Oncol. 2006;7(1):77–82.CrossRefPubMed
25.
go back to reference Chu YW, Yang PC, Yang SC, Shyu YC, Hendrix MJ, Wu R, Wu CW. Selection of invasive and metastatic subpopulations from a human lung adenocarcinoma cell line. Am J Respir Cell Mol Biol. 1997;17(3):353–60.CrossRefPubMed Chu YW, Yang PC, Yang SC, Shyu YC, Hendrix MJ, Wu R, Wu CW. Selection of invasive and metastatic subpopulations from a human lung adenocarcinoma cell line. Am J Respir Cell Mol Biol. 1997;17(3):353–60.CrossRefPubMed
26.
go back to reference Chang K-T, Tsai M-J, Cheng Y-T, Chen J-J, Hsia R-H, Lo Y-S, Ma Y-R, Weng C-F. Comparative atomic force and scanning electron microscopy: an investigation of structural differentiation of hepatic stellate cells. J Struct Biol. 2009;167(3):200–8.CrossRefPubMed Chang K-T, Tsai M-J, Cheng Y-T, Chen J-J, Hsia R-H, Lo Y-S, Ma Y-R, Weng C-F. Comparative atomic force and scanning electron microscopy: an investigation of structural differentiation of hepatic stellate cells. J Struct Biol. 2009;167(3):200–8.CrossRefPubMed
27.
go back to reference Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB. Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci U S A. 1996;93(18):9821–6.CrossRefPubMedPubMedCentral Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB. Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci U S A. 1996;93(18):9821–6.CrossRefPubMedPubMedCentral
28.
go back to reference Chen L-H, Liu D-W, Chang J-L, Chen P-R, Hsu L-P, Lin H-Y, Chou Y-F, Lee C-F, Yang M-C, Wen Y-H. Methylation status of insulin-like growth factor-binding protein 7 concurs with the malignance of oral tongue cancer. J Exp Clin Cancer Res. 2015;34(1):1.CrossRef Chen L-H, Liu D-W, Chang J-L, Chen P-R, Hsu L-P, Lin H-Y, Chou Y-F, Lee C-F, Yang M-C, Wen Y-H. Methylation status of insulin-like growth factor-binding protein 7 concurs with the malignance of oral tongue cancer. J Exp Clin Cancer Res. 2015;34(1):1.CrossRef
29.
go back to reference Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative CT method. Nat Protoc. 2008;3(6):1101–8.CrossRefPubMed Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative CT method. Nat Protoc. 2008;3(6):1101–8.CrossRefPubMed
30.
go back to reference Boyer B, Thiery JP. Epithelium‐mesenchyme interconversion as example of epithelial plasticity. APMIS. 1993;101(1–6):257–68.CrossRefPubMed Boyer B, Thiery JP. Epithelium‐mesenchyme interconversion as example of epithelial plasticity. APMIS. 1993;101(1–6):257–68.CrossRefPubMed
31.
go back to reference Hay E. An overview of epithelio-mesenchymal transformation. Cells Tissues Organs. 1995;154(1):8–20.CrossRef Hay E. An overview of epithelio-mesenchymal transformation. Cells Tissues Organs. 1995;154(1):8–20.CrossRef
32.
go back to reference Machesky LM. Lamellipodia and filopodia in metastasis and invasion. FEBS Lett. 2008;582(14):2102–11.CrossRefPubMed Machesky LM. Lamellipodia and filopodia in metastasis and invasion. FEBS Lett. 2008;582(14):2102–11.CrossRefPubMed
33.
go back to reference Ibarra N, Pollitt A, Insall R. Regulation of actin assembly by SCAR/WAVE proteins. Biochem Soc Trans. 2005;33(Pt 6):1243–6.CrossRefPubMed Ibarra N, Pollitt A, Insall R. Regulation of actin assembly by SCAR/WAVE proteins. Biochem Soc Trans. 2005;33(Pt 6):1243–6.CrossRefPubMed
34.
go back to reference Innocenti M, Zucconi A, Disanza A, Frittoli E, Areces LB, Steffen A, Stradal TE, Di Fiore PP, Carlier M-F, Scita G. Abi1 is essential for the formation and activation of a WAVE2 signalling complex. Nat Cell Biol. 2004;6(4):319–27.CrossRefPubMed Innocenti M, Zucconi A, Disanza A, Frittoli E, Areces LB, Steffen A, Stradal TE, Di Fiore PP, Carlier M-F, Scita G. Abi1 is essential for the formation and activation of a WAVE2 signalling complex. Nat Cell Biol. 2004;6(4):319–27.CrossRefPubMed
35.
go back to reference Kunda P, Craig G, Dominguez V, Baum B. Abi, Sra1, and Kette control the stability and localization of SCAR/WAVE to regulate the formation of actin-based protrusions. Curr Biol. 2003;13(21):1867–75.CrossRefPubMed Kunda P, Craig G, Dominguez V, Baum B. Abi, Sra1, and Kette control the stability and localization of SCAR/WAVE to regulate the formation of actin-based protrusions. Curr Biol. 2003;13(21):1867–75.CrossRefPubMed
36.
go back to reference Svitkina TM, Bulanova EA, Chaga OY, Vignjevic DM, Kojima S-I, Vasiliev JM, Borisy GG. Mechanism of filopodia initiation by reorganization of a dendritic network. J Cell Biol. 2003;160(3):409–21.CrossRefPubMedPubMedCentral Svitkina TM, Bulanova EA, Chaga OY, Vignjevic DM, Kojima S-I, Vasiliev JM, Borisy GG. Mechanism of filopodia initiation by reorganization of a dendritic network. J Cell Biol. 2003;160(3):409–21.CrossRefPubMedPubMedCentral
37.
go back to reference Hashimoto Y, Parsons M, Adams JC. Dual actin-bundling and protein kinase C-binding activities of fascin regulate carcinoma cell migration downstream of Rac and contribute to metastasis. Mol Biol Cell. 2007;18(11):4591–602.CrossRefPubMedPubMedCentral Hashimoto Y, Parsons M, Adams JC. Dual actin-bundling and protein kinase C-binding activities of fascin regulate carcinoma cell migration downstream of Rac and contribute to metastasis. Mol Biol Cell. 2007;18(11):4591–602.CrossRefPubMedPubMedCentral
38.
39.
go back to reference Roll JD, Rivenbark AG, Jones WD, Coleman WB. DNMT3b overexpression contributes to a hypermethylator phenotype in human breast cancer cell lines. Mol Cancer. 2008;7(1):15.CrossRefPubMedPubMedCentral Roll JD, Rivenbark AG, Jones WD, Coleman WB. DNMT3b overexpression contributes to a hypermethylator phenotype in human breast cancer cell lines. Mol Cancer. 2008;7(1):15.CrossRefPubMedPubMedCentral
40.
go back to reference Kanai Y, Ushijima S, Kondo Y, Nakanishi Y, Hirohashi S. DNA methyltransferase expression and DNA methylation of CPG islands and peri‐centromeric satellite regions in human colorectal and stomach cancers. Int J Cancer. 2001;91(2):205–12.CrossRefPubMed Kanai Y, Ushijima S, Kondo Y, Nakanishi Y, Hirohashi S. DNA methyltransferase expression and DNA methylation of CPG islands and peri‐centromeric satellite regions in human colorectal and stomach cancers. Int J Cancer. 2001;91(2):205–12.CrossRefPubMed
41.
go back to reference Nosho K, Shima K, Irahara N, Kure S, Baba Y, Kirkner GJ, Chen L, Gokhale S, Hazra A, Spiegelman D. DNMT3B expression might contribute to CpG island methylator phenotype in colorectal cancer. Clin Cancer Res. 2009;15(11):3663–71.CrossRefPubMedPubMedCentral Nosho K, Shima K, Irahara N, Kure S, Baba Y, Kirkner GJ, Chen L, Gokhale S, Hazra A, Spiegelman D. DNMT3B expression might contribute to CpG island methylator phenotype in colorectal cancer. Clin Cancer Res. 2009;15(11):3663–71.CrossRefPubMedPubMedCentral
42.
go back to reference Lin T-S, Lee H, Chen R-A, Ho M-L, Lin C-Y, Chen Y-H, Tsai YY, Chou M-C, Cheng Y-W. An association of DNMT3b protein expression with P16INK4a promoter hypermethylation in non-smoking female lung cancer with human papillomavirus infection. Cancer Lett. 2005;226(1):77–84.CrossRefPubMed Lin T-S, Lee H, Chen R-A, Ho M-L, Lin C-Y, Chen Y-H, Tsai YY, Chou M-C, Cheng Y-W. An association of DNMT3b protein expression with P16INK4a promoter hypermethylation in non-smoking female lung cancer with human papillomavirus infection. Cancer Lett. 2005;226(1):77–84.CrossRefPubMed
43.
go back to reference Lin H, Yamada Y, Nguyen S, Linhart H, Jackson-Grusby L, Meissner A, Meletis K, Lo G, Jaenisch R. Suppression of intestinal neoplasia by deletion of Dnmt3b. Mol Cell Biol. 2006;26(8):2976–83.CrossRefPubMedPubMedCentral Lin H, Yamada Y, Nguyen S, Linhart H, Jackson-Grusby L, Meissner A, Meletis K, Lo G, Jaenisch R. Suppression of intestinal neoplasia by deletion of Dnmt3b. Mol Cell Biol. 2006;26(8):2976–83.CrossRefPubMedPubMedCentral
44.
go back to reference Linhart HG, Lin H, Yamada Y, Moran E, Steine EJ, Gokhale S, Lo G, Cantu E, Ehrich M, He T. Dnmt3b promotes tumorigenesis in vivo by gene-specific de novo methylation and transcriptional silencing. Genes Dev. 2007;21(23):3110–22.CrossRefPubMedPubMedCentral Linhart HG, Lin H, Yamada Y, Moran E, Steine EJ, Gokhale S, Lo G, Cantu E, Ehrich M, He T. Dnmt3b promotes tumorigenesis in vivo by gene-specific de novo methylation and transcriptional silencing. Genes Dev. 2007;21(23):3110–22.CrossRefPubMedPubMedCentral
45.
go back to reference Hameed R, Raimondi SL. The role of aberrant DNMT3Bs in tumor progression–a review. Cancer Cell Microenviron. 2015;2(2):e847. 10-14800/ccm. 14847. Hameed R, Raimondi SL. The role of aberrant DNMT3Bs in tumor progression–a review. Cancer Cell Microenviron. 2015;2(2):e847. 10-14800/ccm. 14847.
46.
go back to reference Ostler K, Davis E, Payne S, Gosalia B, Exposito-Cespedes J, Le Beau M, Godley L. Cancer cells express aberrant DNMT3B transcripts encoding truncated proteins. Oncogene. 2007;26(38):5553–63.CrossRefPubMedPubMedCentral Ostler K, Davis E, Payne S, Gosalia B, Exposito-Cespedes J, Le Beau M, Godley L. Cancer cells express aberrant DNMT3B transcripts encoding truncated proteins. Oncogene. 2007;26(38):5553–63.CrossRefPubMedPubMedCentral
47.
go back to reference Brambert PR, Kelpsch DJ, Hameed R, Desai CV, Calafiore G, Godley LA, Raimondi SL. DNMT3B7 Expression Promotes Tumor Progression to a More Aggressive Phenotype in Breast Cancer Cells. PLoS ONE. 2015;10(1):e0117310.CrossRefPubMedPubMedCentral Brambert PR, Kelpsch DJ, Hameed R, Desai CV, Calafiore G, Godley LA, Raimondi SL. DNMT3B7 Expression Promotes Tumor Progression to a More Aggressive Phenotype in Breast Cancer Cells. PLoS ONE. 2015;10(1):e0117310.CrossRefPubMedPubMedCentral
48.
go back to reference Fabbri M, Garzon R, Cimmino A, Liu Z, Zanesi N, Callegari E, Liu S, Alder H, Costinean S, Fernandez-Cymering C. MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferases 3A and 3B. Proc Natl Acad Sci. 2007;104(40):15805–10.CrossRefPubMedPubMedCentral Fabbri M, Garzon R, Cimmino A, Liu Z, Zanesi N, Callegari E, Liu S, Alder H, Costinean S, Fernandez-Cymering C. MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferases 3A and 3B. Proc Natl Acad Sci. 2007;104(40):15805–10.CrossRefPubMedPubMedCentral
49.
go back to reference Cortez MA, Nicoloso MS, Shimizu M, Rossi S, Gopisetty G, Molina JR, Carlotti C, Tirapelli D, Neder L, Brassesco MS. miR‐29b and miR‐125a regulate podoplanin and suppress invasion in glioblastoma. Genes Chromosom Cancer. 2010;49(11):981–90.CrossRefPubMed Cortez MA, Nicoloso MS, Shimizu M, Rossi S, Gopisetty G, Molina JR, Carlotti C, Tirapelli D, Neder L, Brassesco MS. miR‐29b and miR‐125a regulate podoplanin and suppress invasion in glioblastoma. Genes Chromosom Cancer. 2010;49(11):981–90.CrossRefPubMed
50.
go back to reference Ru P, Steele R, Newhall P, Phillips NJ, Toth K, Ray RB. miRNA-29b suppresses prostate cancer metastasis by regulating epithelial–mesenchymal transition signaling. Mol Cancer Ther. 2012;11(5):1166–73.CrossRefPubMed Ru P, Steele R, Newhall P, Phillips NJ, Toth K, Ray RB. miRNA-29b suppresses prostate cancer metastasis by regulating epithelial–mesenchymal transition signaling. Mol Cancer Ther. 2012;11(5):1166–73.CrossRefPubMed
51.
go back to reference Flavin R, Smyth P, Barrett C, Russell S, Wen H, Wei J, Laios A, O’Toole S, Ring M, Denning K. miR-29b expression is associated with disease-free survival in patients with ovarian serous carcinoma. Int J Gynecol Cancer. 2009;19(4):641–7.CrossRefPubMed Flavin R, Smyth P, Barrett C, Russell S, Wen H, Wei J, Laios A, O’Toole S, Ring M, Denning K. miR-29b expression is associated with disease-free survival in patients with ovarian serous carcinoma. Int J Gynecol Cancer. 2009;19(4):641–7.CrossRefPubMed
52.
go back to reference Kinoshita T, Nohata N, Hanazawa T, Kikkawa N, Yamamoto N, Yoshino H, Itesako T, Enokida H, Nakagawa M, Okamoto Y. Tumour-suppressive microRNA-29 s inhibit cancer cell migration and invasion by targeting laminin–integrin signalling in head and neck squamous cell carcinoma. Br J Cancer. 2013;109(10):2636–45.CrossRefPubMedPubMedCentral Kinoshita T, Nohata N, Hanazawa T, Kikkawa N, Yamamoto N, Yoshino H, Itesako T, Enokida H, Nakagawa M, Okamoto Y. Tumour-suppressive microRNA-29 s inhibit cancer cell migration and invasion by targeting laminin–integrin signalling in head and neck squamous cell carcinoma. Br J Cancer. 2013;109(10):2636–45.CrossRefPubMedPubMedCentral
53.
go back to reference Xiong Y, Fang JH, Yun JP, Yang J, Zhang Y, Jia WH, Zhuang SM. Effects of MicroRNA‐29 on apoptosis, tumorigenicity, and prognosis of hepatocellular carcinoma. Hepatology. 2010;51(3):836–45.PubMed Xiong Y, Fang JH, Yun JP, Yang J, Zhang Y, Jia WH, Zhuang SM. Effects of MicroRNA‐29 on apoptosis, tumorigenicity, and prognosis of hepatocellular carcinoma. Hepatology. 2010;51(3):836–45.PubMed
55.
go back to reference Garzon R, Heaphy CE, Havelange V, Fabbri M, Volinia S, Tsao T, Zanesi N, Kornblau SM, Marcucci G, Calin GA. MicroRNA 29b functions in acute myeloid leukemia. Blood. 2009;114(26):5331–41.CrossRefPubMedPubMedCentral Garzon R, Heaphy CE, Havelange V, Fabbri M, Volinia S, Tsao T, Zanesi N, Kornblau SM, Marcucci G, Calin GA. MicroRNA 29b functions in acute myeloid leukemia. Blood. 2009;114(26):5331–41.CrossRefPubMedPubMedCentral
56.
go back to reference Rothschild SI, Tschan M, Federzoni E, Jaggi R, Fey M, Gugger M, Gautschi O. MicroRNA-29b is involved in the Src-ID1 signaling pathway and is dysregulated in human lung adenocarcinoma. Oncogene. 2012;31(38):4221–32.CrossRefPubMed Rothschild SI, Tschan M, Federzoni E, Jaggi R, Fey M, Gugger M, Gautschi O. MicroRNA-29b is involved in the Src-ID1 signaling pathway and is dysregulated in human lung adenocarcinoma. Oncogene. 2012;31(38):4221–32.CrossRefPubMed
57.
go back to reference Li H, Solomon E, Muggy SD, Sun D, Zolkiewska A. Metalloprotease-disintegrin ADAM12 expression is regulated by Notch signaling via microRNA-29. J Biol Chem. 2011;286(24):21500–10.CrossRefPubMedPubMedCentral Li H, Solomon E, Muggy SD, Sun D, Zolkiewska A. Metalloprotease-disintegrin ADAM12 expression is regulated by Notch signaling via microRNA-29. J Biol Chem. 2011;286(24):21500–10.CrossRefPubMedPubMedCentral
58.
go back to reference Chou J, Lin JH, Brenot A, Kim J-W, Provot S, Werb Z. GATA3 suppresses metastasis and modulates the tumour microenvironment by regulating microRNA-29b expression. Nat Cell Biol. 2013;15(2):201–13.CrossRefPubMedPubMedCentral Chou J, Lin JH, Brenot A, Kim J-W, Provot S, Werb Z. GATA3 suppresses metastasis and modulates the tumour microenvironment by regulating microRNA-29b expression. Nat Cell Biol. 2013;15(2):201–13.CrossRefPubMedPubMedCentral
59.
go back to reference Melo SA, Kalluri R. miR-29b moulds the tumour microenvironment to repress metastasis. Nat Cell Biol. 2013;15(2):139–40.CrossRefPubMed Melo SA, Kalluri R. miR-29b moulds the tumour microenvironment to repress metastasis. Nat Cell Biol. 2013;15(2):139–40.CrossRefPubMed
60.
go back to reference Garzon R, Liu S, Fabbri M, Liu Z, Heaphy CE, Callegari E, Schwind S, Pang J, Yu J, Muthusamy N. 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, Schwind S, Pang J, Yu J, Muthusamy N. 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
61.
63.
go back to reference Polyak K, Xia Y, Zweier JL, Kinzler KW, Vogelstein B. A model for p53-induced apoptosis. Nature. 1997;389(6648):300–5.CrossRefPubMed Polyak K, Xia Y, Zweier JL, Kinzler KW, Vogelstein B. A model for p53-induced apoptosis. Nature. 1997;389(6648):300–5.CrossRefPubMed
64.
go back to reference Liu Y, Borchert GL, Donald SP, Diwan BA, Anver M, Phang JM. Proline oxidase functions as a mitochondrial tumor suppressor in human cancers. Cancer Res. 2009;69(16):6414–22.CrossRefPubMedPubMedCentral Liu Y, Borchert GL, Donald SP, Diwan BA, Anver M, Phang JM. Proline oxidase functions as a mitochondrial tumor suppressor in human cancers. Cancer Res. 2009;69(16):6414–22.CrossRefPubMedPubMedCentral
65.
go back to reference Liu W, Zabirnyk O, Wang H, Shiao Y, Nickerson M, Khalil S, Anderson L, Perantoni A, Phang J. miR-23b* targets proline oxidase, a novel tumor suppressor protein in renal cancer. Oncogene. 2010;29(35):4914–24.CrossRefPubMedPubMedCentral Liu W, Zabirnyk O, Wang H, Shiao Y, Nickerson M, Khalil S, Anderson L, Perantoni A, Phang J. miR-23b* targets proline oxidase, a novel tumor suppressor protein in renal cancer. Oncogene. 2010;29(35):4914–24.CrossRefPubMedPubMedCentral
67.
go back to reference Moustakas A, Heldin CH. Signaling networks guiding epithelial–mesenchymal transitions during embryogenesis and cancer progression. Cancer Sci. 2007;98(10):1512–20.CrossRefPubMed Moustakas A, Heldin CH. Signaling networks guiding epithelial–mesenchymal transitions during embryogenesis and cancer progression. Cancer Sci. 2007;98(10):1512–20.CrossRefPubMed
68.
go back to reference Zhang Y, Feng X-H, Wu R-Y, Derynck R. Receptor-associated Mad homologues synergize as effectors of the TGF-β response. Nature. 1996;383:168–72.CrossRefPubMed Zhang Y, Feng X-H, Wu R-Y, Derynck R. Receptor-associated Mad homologues synergize as effectors of the TGF-β response. Nature. 1996;383:168–72.CrossRefPubMed
69.
go back to reference Yang L, Mao C, Teng Y, Li W, Zhang J, Cheng X, Li X, Han X, Xia Z, Deng H. Targeted disruption of Smad4 in mouse epidermis results in failure of hair follicle cycling and formation of skin tumors. Cancer Res. 2005;65(19):8671–8.CrossRefPubMed Yang L, Mao C, Teng Y, Li W, Zhang J, Cheng X, Li X, Han X, Xia Z, Deng H. Targeted disruption of Smad4 in mouse epidermis results in failure of hair follicle cycling and formation of skin tumors. Cancer Res. 2005;65(19):8671–8.CrossRefPubMed
70.
go back to reference Qiao W, Li A, Owens P, Xu X, Wang X, Deng C. Hair follicle defects and squamous cell carcinoma formation in Smad4 conditional knockout mouse skin. Oncogene. 2006;25(2):207–17.PubMed Qiao W, Li A, Owens P, Xu X, Wang X, Deng C. Hair follicle defects and squamous cell carcinoma formation in Smad4 conditional knockout mouse skin. Oncogene. 2006;25(2):207–17.PubMed
71.
go back to reference Bornstein S, White R, Malkoski S, Oka M, Han G, Cleaver T, Reh D, Andersen P, Gross N, Olson S. Smad4 loss in mice causes spontaneous head and neck cancer with increased genomic instability and inflammation. J Clin Invest. 2009;119(11):3408.PubMedPubMedCentral Bornstein S, White R, Malkoski S, Oka M, Han G, Cleaver T, Reh D, Andersen P, Gross N, Olson S. Smad4 loss in mice causes spontaneous head and neck cancer with increased genomic instability and inflammation. J Clin Invest. 2009;119(11):3408.PubMedPubMedCentral
72.
go back to reference Agrawal N, Frederick MJ, Pickering CR, Bettegowda C, Chang K, Li RJ,Fakhry C, Xie T-X, Zhang J, Wang J. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science. 2011;333(6046):1154–7.CrossRefPubMedPubMedCentral Agrawal N, Frederick MJ, Pickering CR, Bettegowda C, Chang K, Li RJ,Fakhry C, Xie T-X, Zhang J, Wang J. Exome sequencing of head and neck squamous cell carcinoma reveals inactivating mutations in NOTCH1. Science. 2011;333(6046):1154–7.CrossRefPubMedPubMedCentral
73.
go back to reference Kim SK, Fan Y, Papadimitrakopoulou V, Clayman G, Hittleman WN, Hong WK, Lotan R, Mao L. DPC4, a candidate tumor suppressor gene, is altered infrequently in head and neck squamous cell carcinoma. Cancer Res. 1996;56(11):2519–21.PubMed Kim SK, Fan Y, Papadimitrakopoulou V, Clayman G, Hittleman WN, Hong WK, Lotan R, Mao L. DPC4, a candidate tumor suppressor gene, is altered infrequently in head and neck squamous cell carcinoma. Cancer Res. 1996;56(11):2519–21.PubMed
74.
go back to reference Cardenas H, Vieth E, Lee J, Segar M, Liu Y, Nephew KP, Matei D. TGF-β induces global changes in DNA methylation during the epithelial-to-mesenchymal transition in ovarian cancer cells. Epigenetics. 2014;9(11):1461–72.CrossRefPubMedPubMedCentral Cardenas H, Vieth E, Lee J, Segar M, Liu Y, Nephew KP, Matei D. TGF-β induces global changes in DNA methylation during the epithelial-to-mesenchymal transition in ovarian cancer cells. Epigenetics. 2014;9(11):1461–72.CrossRefPubMedPubMedCentral
76.
go back to reference Ozdamar B, Bose R, Barrios-Rodiles M, Wang H-R, Zhang Y, Wrana JL. Regulation of the polarity protein Par6 by TGFß receptors controls epithelial cell plasticity. Science. 2005;307(5715):1603–9.CrossRefPubMed Ozdamar B, Bose R, Barrios-Rodiles M, Wang H-R, Zhang Y, Wrana JL. Regulation of the polarity protein Par6 by TGFß receptors controls epithelial cell plasticity. Science. 2005;307(5715):1603–9.CrossRefPubMed
77.
go back to reference Yamashita M, Fatyol K, Jin C, Wang X, Liu Z, Zhang YE. TRAF6 mediates Smad-independent activation of JNK and p38 by TGF-β. Mol Cell. 2008;31(6):918–24.CrossRefPubMedPubMedCentral Yamashita M, Fatyol K, Jin C, Wang X, Liu Z, Zhang YE. TRAF6 mediates Smad-independent activation of JNK and p38 by TGF-β. Mol Cell. 2008;31(6):918–24.CrossRefPubMedPubMedCentral
79.
go back to reference Grinnell F, Billingham RE, Burgess L. Distribution of fibronectin during wound healing in vivo. J Investig Dermatol. 1981;76(3):181–9.CrossRefPubMed Grinnell F, Billingham RE, Burgess L. Distribution of fibronectin during wound healing in vivo. J Investig Dermatol. 1981;76(3):181–9.CrossRefPubMed
80.
go back to reference Han S, Khuri FR, Roman J. Fibronectin stimulates non–small cell lung carcinoma cell growth through activation of Akt/mammalian target of rapamycin/S6 kinase and inactivation of LKB1/AMP-activated protein kinase signal pathways. Cancer Res. 2006;66(1):315–23.CrossRefPubMed Han S, Khuri FR, Roman J. Fibronectin stimulates non–small cell lung carcinoma cell growth through activation of Akt/mammalian target of rapamycin/S6 kinase and inactivation of LKB1/AMP-activated protein kinase signal pathways. Cancer Res. 2006;66(1):315–23.CrossRefPubMed
81.
go back to reference Park J, Schwarzbauer JE. Mammary epithelial cell interactions with fibronectin stimulate epithelial-mesenchymal transition. Oncogene. 2014;33(13):1649–57.CrossRefPubMed Park J, Schwarzbauer JE. Mammary epithelial cell interactions with fibronectin stimulate epithelial-mesenchymal transition. Oncogene. 2014;33(13):1649–57.CrossRefPubMed
82.
go back to reference Chen S-Y, Lin J-S, Yang B-C. Modulation of tumor cell stiffness and migration by type IV collagen through direct activation of integrin signaling pathway. Arch Biochem Biophys. 2014;555:1–8.CrossRefPubMed Chen S-Y, Lin J-S, Yang B-C. Modulation of tumor cell stiffness and migration by type IV collagen through direct activation of integrin signaling pathway. Arch Biochem Biophys. 2014;555:1–8.CrossRefPubMed
83.
go back to reference Jinnin M, Ihn H, Asano Y, Yamane K, Trojanowska M, Tamaki K. Tenascin-C upregulation by transforming growth factor-β in human dermal fibroblasts involves Smad3, Sp1, and Ets1. Oncogene. 2004;23(9):1656–67.CrossRefPubMed Jinnin M, Ihn H, Asano Y, Yamane K, Trojanowska M, Tamaki K. Tenascin-C upregulation by transforming growth factor-β in human dermal fibroblasts involves Smad3, Sp1, and Ets1. Oncogene. 2004;23(9):1656–67.CrossRefPubMed
84.
go back to reference Jahkola T, Toivonen T, Virtanen I, von Smitten K, Nordling S, von Boguslawski K, Haglund C, Nevanlinna H, Blomqvist C. Tenascin-C expression in invasion border of early breast cancer: a predictor of local and distant recurrence. Br J Cancer. 1998;78(11):1507.CrossRefPubMedPubMedCentral Jahkola T, Toivonen T, Virtanen I, von Smitten K, Nordling S, von Boguslawski K, Haglund C, Nevanlinna H, Blomqvist C. Tenascin-C expression in invasion border of early breast cancer: a predictor of local and distant recurrence. Br J Cancer. 1998;78(11):1507.CrossRefPubMedPubMedCentral
85.
go back to reference Orend G, Chiquet-Ehrismann R. Tenascin-C induced signaling in cancer. Cancer Lett. 2006;244(2):143–63.CrossRefPubMed Orend G, Chiquet-Ehrismann R. Tenascin-C induced signaling in cancer. Cancer Lett. 2006;244(2):143–63.CrossRefPubMed
86.
go back to reference De Wever O, Nguyen Q-D, Van Hoorde L, Bracke M, Bruyneel E, Gespach C, Mareel M. Tenascin-C and SF/HGF produced by myofibroblasts in vitro provide convergent pro-invasive signals to human colon cancer cells through RhoA and Rac. FASEB J. 2004;18(9):1016–8.PubMed De Wever O, Nguyen Q-D, Van Hoorde L, Bracke M, Bruyneel E, Gespach C, Mareel M. Tenascin-C and SF/HGF produced by myofibroblasts in vitro provide convergent pro-invasive signals to human colon cancer cells through RhoA and Rac. FASEB J. 2004;18(9):1016–8.PubMed
87.
go back to reference Dandachi N, Hauser‐Kronberger C, More E, Wiesener B, Hacker G, Dietze O, Wirl G. Co‐expression of tenascin‐C and vimentin in human breast cancer cells indicates phenotypic transdifferentiation during tumour progression: correlation with histopathological parameters, hormone receptors, and oncoproteins. J Pathol. 2001;193(2):181–9.CrossRefPubMed Dandachi N, Hauser‐Kronberger C, More E, Wiesener B, Hacker G, Dietze O, Wirl G. Co‐expression of tenascin‐C and vimentin in human breast cancer cells indicates phenotypic transdifferentiation during tumour progression: correlation with histopathological parameters, hormone receptors, and oncoproteins. J Pathol. 2001;193(2):181–9.CrossRefPubMed
Metadata
Title
Involvement of DNMT 3B promotes epithelial-mesenchymal transition and gene expression profile of invasive head and neck squamous cell carcinomas cell lines
Authors
Li-Hsuen Chen
Wen-Lin Hsu
Yen-Ju Tseng
Dai-Wei Liu
Ching-Feng Weng
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/s12885-016-2468-x

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