Skip to main content
Top
Published in: BMC Cancer 1/2015

Open Access 01-12-2015 | Research article

Ras induces experimental lung metastasis through up-regulation of RbAp46 to suppress RECK promoter activity

Authors: Hsuan-Heng Yeh, Yu-Fen Tseng, Yu-Chiao Hsu, Sheng-Hui Lan, Shan-Ying Wu, Giri Raghavaraju, Da-En Cheng, Ying-Ray Lee, Tsuey-Yu Chang, Nan-Haw Chow, Wen-Chun Hung, Hsiao-Sheng Liu

Published in: BMC Cancer | Issue 1/2015

Login to get access

Abstract

Background

Mutant Ras plays multiple functions in tumorigenesis including tumor formation and metastasis. Reversion-inducing cysteine-rich protein with Kazal motifs (RECK), a metastasis inhibitor gene, suppresses matrix metalloproteinase (MMP) activity in the metastatic cascade. Clarifying the relationship between Ras and RECK and understanding the underlying molecular mechanism may lead to the development of better treatment for Ras-related tumors.

Methods

Suppression subtractive hybridization PCR (SSH PCR) was conducted to identify Ha-rasval12 up-regulated genes in bladder cancer cells. Stable cell lines of human breast cancer (MCF-7-ras) and mouse NIH3T3 fibroblasts (7–4) harboring the inducible Ha-rasval12 oncogene, which could be induced by isopropylthio-β-D-galactoside (IPTG), were used to clarify the relationship between Ras and the up-regulated genes. Chromatin immunoprecipitation (ChIP) assay, DNA affinity precipitation assay (DAPA) and RECK reporter gene assay were utilized to confirm the complex formation and binding with promoters.

Results

Retinoblastoma binding protein-7 (RbAp46) was identified and confirmed as a Ha-rasval12 up-regulated gene. RbAp46 could bind with histone deacetylase (HDAC1) and Sp1, followed by binding to RECK promoter at the Sp1 site resulting in repression of RECK expression. High expression of Ras protein accompanied with high RbAp46 and low RECK expression were detected in 75% (3/4) of the clinical bladder cancer tumor tissues compared to the adjacent normal parts. Ras induced RbAp46 expression increases invasion of the bladder cancer T24 cells and MMP-9 activity was increased, which was confirmed by specific lentiviral shRNAs inhibitors against Ras and RbAp46. Similarly, knockdown of RbAp46 expression in the stable NIH3T3 cells “7-4” by shRNA decreased Ras-related lung metastasis using a xenograft nude mice model.

Conclusions

We confirmed that RbAp46 is a Ha-rasval12 up-regulated gene and binds with HDAC1 and Sp1. Furthermore, RbAp46 binds to the RECK promoter at the Sp1 site via recruitment by Sp1. RECK is subsequently activated, leading to increased MMP9 activity, which may lead to increased metastasis in vivo. Our findings of Ras upregulation of RbAp46 may lead to revealing a novel mechanism of Ras-related tumor cell metastasis.
Appendix
Available only for authorised users
Literature
1.
go back to reference Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003;3(1):11–22.CrossRefPubMed Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003;3(1):11–22.CrossRefPubMed
2.
go back to reference Malumbres M, Barbacid M. RAS oncogenes: the first 30 years. Nat Rev. 2003;3(6):459–65.CrossRef Malumbres M, Barbacid M. RAS oncogenes: the first 30 years. Nat Rev. 2003;3(6):459–65.CrossRef
3.
go back to reference Shields JM, Pruitt K, McFall A, Shaub A, Der CJ. Understanding Ras: ‘it ain’t over ’til it’s over’. Trends Cell Biol. 2000;10(4):147–54.CrossRefPubMed Shields JM, Pruitt K, McFall A, Shaub A, Der CJ. Understanding Ras: ‘it ain’t over ’til it’s over’. Trends Cell Biol. 2000;10(4):147–54.CrossRefPubMed
4.
go back to reference Campbell PM, Der CJ. Oncogenic Ras and its role in tumor cell invasion and metastasis. Semin Cancer Biol. 2004;14(2):105–14.CrossRefPubMed Campbell PM, Der CJ. Oncogenic Ras and its role in tumor cell invasion and metastasis. Semin Cancer Biol. 2004;14(2):105–14.CrossRefPubMed
5.
go back to reference Takahashi C, Sheng Z, Horan TP, Kitayama H, Maki M, Hitomi K, et al. Regulation of matrix metalloproteinase-9 and inhibition of tumor invasion by the membrane-anchored glycoprotein RECK. Proc Natl Acad Sci U S A. 1998;95(22):13221–6.CrossRefPubMedPubMedCentral Takahashi C, Sheng Z, Horan TP, Kitayama H, Maki M, Hitomi K, et al. Regulation of matrix metalloproteinase-9 and inhibition of tumor invasion by the membrane-anchored glycoprotein RECK. Proc Natl Acad Sci U S A. 1998;95(22):13221–6.CrossRefPubMedPubMedCentral
6.
go back to reference Oh J, Takahashi R, Kondo S, Mizoguchi A, Adachi E, Sasahara RM, et al. The membrane-anchored MMP inhibitor RECK is a key regulator of extracellular matrix integrity and angiogenesis. Cell. 2001;107(6):789–800.CrossRefPubMed Oh J, Takahashi R, Kondo S, Mizoguchi A, Adachi E, Sasahara RM, et al. The membrane-anchored MMP inhibitor RECK is a key regulator of extracellular matrix integrity and angiogenesis. Cell. 2001;107(6):789–800.CrossRefPubMed
7.
go back to reference Clark JC, Thomas DM, Choong PF, Dass CR. RECK–a newly discovered inhibitor of metastasis with prognostic significance in multiple forms of cancer. Cancer Metastasis Rev. 2007;26(3–4):675–83.CrossRefPubMed Clark JC, Thomas DM, Choong PF, Dass CR. RECK–a newly discovered inhibitor of metastasis with prognostic significance in multiple forms of cancer. Cancer Metastasis Rev. 2007;26(3–4):675–83.CrossRefPubMed
8.
go back to reference Sasahara RM, Takahashi C, Sogayar MC, Noda M. Oncogene-mediated downregulation of RECK, a novel transformation suppressor gene. Braz J Med Biol Res. 1999;32(7):891–5.CrossRefPubMed Sasahara RM, Takahashi C, Sogayar MC, Noda M. Oncogene-mediated downregulation of RECK, a novel transformation suppressor gene. Braz J Med Biol Res. 1999;32(7):891–5.CrossRefPubMed
9.
go back to reference Jeon HW, Lee YM. Inhibition of histone deacetylase attenuates hypoxia-induced migration and invasion of cancer cells via the restoration of RECK expression. Mol Cancer Ther. 2010;9(5):1361–70.CrossRefPubMed Jeon HW, Lee YM. Inhibition of histone deacetylase attenuates hypoxia-induced migration and invasion of cancer cells via the restoration of RECK expression. Mol Cancer Ther. 2010;9(5):1361–70.CrossRefPubMed
10.
go back to reference Chang HC, Cho CY, Hung WC. Silencing of the metastasis suppressor RECK by RAS oncogene is mediated by DNA methyltransferase 3b-induced promoter methylation. Cancer Res. 2006;66(17):8413–20.CrossRefPubMed Chang HC, Cho CY, Hung WC. Silencing of the metastasis suppressor RECK by RAS oncogene is mediated by DNA methyltransferase 3b-induced promoter methylation. Cancer Res. 2006;66(17):8413–20.CrossRefPubMed
11.
go back to reference Loayza-Puch F, Yoshida Y, Matsuzaki T, Takahashi C, Kitayama H, Noda M. Hypoxia and RAS-signaling pathways converge on, and cooperatively downregulate, the RECK tumor-suppressor protein through microRNAs. Oncogene. 2010;29(18):2638–48.CrossRefPubMed Loayza-Puch F, Yoshida Y, Matsuzaki T, Takahashi C, Kitayama H, Noda M. Hypoxia and RAS-signaling pathways converge on, and cooperatively downregulate, the RECK tumor-suppressor protein through microRNAs. Oncogene. 2010;29(18):2638–48.CrossRefPubMed
12.
go back to reference Sasahara RM, Takahashi C, Noda M. Involvement of the Sp1 site in ras-mediated downregulation of the RECK metastasis suppressor gene. Biochem Biophys Res Commun. 1999;264(3):668–75.CrossRefPubMed Sasahara RM, Takahashi C, Noda M. Involvement of the Sp1 site in ras-mediated downregulation of the RECK metastasis suppressor gene. Biochem Biophys Res Commun. 1999;264(3):668–75.CrossRefPubMed
13.
go back to reference Chang HC, Liu LT, Hung WC. Involvement of histone deacetylation in ras-induced down-regulation of the metastasis suppressor RECK. Cell Signal. 2004;16(6):675–9.CrossRefPubMed Chang HC, Liu LT, Hung WC. Involvement of histone deacetylation in ras-induced down-regulation of the metastasis suppressor RECK. Cell Signal. 2004;16(6):675–9.CrossRefPubMed
14.
go back to reference Huang S, Lee WH, Lee EY. A cellular protein that competes with SV40 T antigen for binding to the retinoblastoma gene product. Nature. 1991;350(6314):160–2.CrossRefPubMed Huang S, Lee WH, Lee EY. A cellular protein that competes with SV40 T antigen for binding to the retinoblastoma gene product. Nature. 1991;350(6314):160–2.CrossRefPubMed
15.
go back to reference Ahringer J. NuRD and SIN3 histone deacetylase complexes in development. Trends Genet. 2000;16(8):351–6.CrossRefPubMed Ahringer J. NuRD and SIN3 histone deacetylase complexes in development. Trends Genet. 2000;16(8):351–6.CrossRefPubMed
16.
go back to reference Verreault A, Kaufman PD, Kobayashi R, Stillman B. Nucleosome assembly by a complex of CAF-1 and acetylated histones H3/H4. Cell. 1996;87(1):95–104.CrossRefPubMed Verreault A, Kaufman PD, Kobayashi R, Stillman B. Nucleosome assembly by a complex of CAF-1 and acetylated histones H3/H4. Cell. 1996;87(1):95–104.CrossRefPubMed
17.
go back to reference Guan LS, Li GC, Chen CC, Liu LQ, Wang ZY. Rb-associated protein 46 (RbAp46) suppresses the tumorigenicity of adenovirus-transformed human embryonic kidney 293 cells. Int J Cancer. 2001;93(3):333–8.CrossRefPubMed Guan LS, Li GC, Chen CC, Liu LQ, Wang ZY. Rb-associated protein 46 (RbAp46) suppresses the tumorigenicity of adenovirus-transformed human embryonic kidney 293 cells. Int J Cancer. 2001;93(3):333–8.CrossRefPubMed
18.
go back to reference Li GC, Guan LS, Wang ZY. Overexpression of RbAp46 facilitates stress-induced apoptosis and suppresses tumorigenicity of neoplastigenic breast epithelial cells. Int J Cancer. 2003;105(6):762–8.CrossRefPubMed Li GC, Guan LS, Wang ZY. Overexpression of RbAp46 facilitates stress-induced apoptosis and suppresses tumorigenicity of neoplastigenic breast epithelial cells. Int J Cancer. 2003;105(6):762–8.CrossRefPubMed
19.
go back to reference Li GC, Wang ZY. Constitutive expression of RbAp46 induces epithelial-mesenchymal transition in mammary epithelial cells. Anticancer Res. 2006;26(5A):3555–60.PubMed Li GC, Wang ZY. Constitutive expression of RbAp46 induces epithelial-mesenchymal transition in mammary epithelial cells. Anticancer Res. 2006;26(5A):3555–60.PubMed
20.
go back to reference Wang CL, Wang CI, Liao PC, Chen CD, Liang Y, Chuang WY, et al. Discovery of retinoblastoma-associated binding protein 46 as a novel prognostic marker for distant metastasis in nonsmall cell lung cancer by combined analysis of cancer cell secretome and pleural effusion proteome. J Proteome Res. 2009;8(10):4428–40.CrossRefPubMed Wang CL, Wang CI, Liao PC, Chen CD, Liang Y, Chuang WY, et al. Discovery of retinoblastoma-associated binding protein 46 as a novel prognostic marker for distant metastasis in nonsmall cell lung cancer by combined analysis of cancer cell secretome and pleural effusion proteome. J Proteome Res. 2009;8(10):4428–40.CrossRefPubMed
21.
go back to reference Fan J, Banerjee D, Stambrook PJ, Bertino JR. Modulation of cytotoxicity of chemotherapeutic drugs by activated H-ras. Biochem Pharmacol. 1997;53(8):1203–9.CrossRefPubMed Fan J, Banerjee D, Stambrook PJ, Bertino JR. Modulation of cytotoxicity of chemotherapeutic drugs by activated H-ras. Biochem Pharmacol. 1997;53(8):1203–9.CrossRefPubMed
22.
go back to reference Liu HS, Scrable H, Villaret DB, Lieberman MA, Stambrook PJ. Control of Ha-ras-mediated mammalian cell transformation by Escherichia coli regulatory elements. Cancer Res. 1992;52(4):983–9.PubMed Liu HS, Scrable H, Villaret DB, Lieberman MA, Stambrook PJ. Control of Ha-ras-mediated mammalian cell transformation by Escherichia coli regulatory elements. Cancer Res. 1992;52(4):983–9.PubMed
23.
go back to reference Hsu MC, Chang HC, Hung WC. HER-2/neu represses the metastasis suppressor RECK via ERK and Sp transcription factors to promote cell invasion. J Biol Chem. 2006;281(8):4718–25.CrossRefPubMed Hsu MC, Chang HC, Hung WC. HER-2/neu represses the metastasis suppressor RECK via ERK and Sp transcription factors to promote cell invasion. J Biol Chem. 2006;281(8):4718–25.CrossRefPubMed
24.
go back to reference Yeh HH, Lai WW, Chen HH, Liu HS, Su WC. Autocrine IL-6-induced Stat3 activation contributes to the pathogenesis of lung adenocarcinoma and malignant pleural effusion. Oncogene. 2006;25(31):4300–9.CrossRefPubMed Yeh HH, Lai WW, Chen HH, Liu HS, Su WC. Autocrine IL-6-induced Stat3 activation contributes to the pathogenesis of lung adenocarcinoma and malignant pleural effusion. Oncogene. 2006;25(31):4300–9.CrossRefPubMed
25.
go back to reference Wells J, Boyd KE, Fry CJ, Bartley SM, Farnham PJ. Target gene specificity of E2F and pocket protein family members in living cells. Mol Cell Biol. 2000;20(16):5797–807.CrossRefPubMedPubMedCentral Wells J, Boyd KE, Fry CJ, Bartley SM, Farnham PJ. Target gene specificity of E2F and pocket protein family members in living cells. Mol Cell Biol. 2000;20(16):5797–807.CrossRefPubMedPubMedCentral
26.
go back to reference Tzeng CC, Liu HS, Li C, Jin YT, Chen RM, Yang WH, et al. Characterization of two urothelium cancer cell lines derived from a blackfoot disease endemic area in Taiwan. Anticancer Res. 1996;16(4A):1797–804.PubMed Tzeng CC, Liu HS, Li C, Jin YT, Chen RM, Yang WH, et al. Characterization of two urothelium cancer cell lines derived from a blackfoot disease endemic area in Taiwan. Anticancer Res. 1996;16(4A):1797–804.PubMed
27.
go back to reference Choi YS, Jeong S. PI3-kinase and PDK-1 regulate HDAC1-mediated transcriptional repression of transcription factor NF-kappaB. Mol Cells. 2005;20(2):241–6.PubMed Choi YS, Jeong S. PI3-kinase and PDK-1 regulate HDAC1-mediated transcriptional repression of transcription factor NF-kappaB. Mol Cells. 2005;20(2):241–6.PubMed
28.
go back to reference Laduron S, Deplus R, Zhou S, Kholmanskikh O, Godelaine D, De Smet C, et al. MAGE-A1 interacts with adaptor SKIP and the deacetylase HDAC1 to repress transcription. Nucleic Acids Res. 2004;32(14):4340–50.CrossRefPubMedPubMedCentral Laduron S, Deplus R, Zhou S, Kholmanskikh O, Godelaine D, De Smet C, et al. MAGE-A1 interacts with adaptor SKIP and the deacetylase HDAC1 to repress transcription. Nucleic Acids Res. 2004;32(14):4340–50.CrossRefPubMedPubMedCentral
29.
go back to reference Chang CK, Hung WC, Chang HC. The Kazal motifs of RECK protein inhibit MMP-9 secretion and activity and reduce metastasis of lung cancer cells in vitro and in vivo. J Cell Mol Med. 2008;12(6B):2781–9.CrossRefPubMedPubMedCentral Chang CK, Hung WC, Chang HC. The Kazal motifs of RECK protein inhibit MMP-9 secretion and activity and reduce metastasis of lung cancer cells in vitro and in vivo. J Cell Mol Med. 2008;12(6B):2781–9.CrossRefPubMedPubMedCentral
30.
go back to reference Hsieh CC, Wu SY, Lan SH, Weng TY, Tsai Y, Liu HS. Effect of Ha-rasval12 on nm23 expression, tumor formation and metastasis of the transformants, and immunomodulation in tumor-bearing mice. Anticancer Res. 2010;30(9):3585–92.PubMed Hsieh CC, Wu SY, Lan SH, Weng TY, Tsai Y, Liu HS. Effect of Ha-rasval12 on nm23 expression, tumor formation and metastasis of the transformants, and immunomodulation in tumor-bearing mice. Anticancer Res. 2010;30(9):3585–92.PubMed
31.
go back to reference Yeh HH, Wu CH, Giri R, Kato K, Kohno K, Izumi H, et al. Oncogenic Ras-induced morphologic change is through MEK/ERK signaling pathway to downregulate Stat3 at a posttranslational level in NIH3T3 cells. Neoplasia. 2008;10(1):52–60.CrossRefPubMedPubMedCentral Yeh HH, Wu CH, Giri R, Kato K, Kohno K, Izumi H, et al. Oncogenic Ras-induced morphologic change is through MEK/ERK signaling pathway to downregulate Stat3 at a posttranslational level in NIH3T3 cells. Neoplasia. 2008;10(1):52–60.CrossRefPubMedPubMedCentral
32.
33.
go back to reference Mazumdar A, Wang RA, Mishra SK, Adam L, Bagheri-Yarmand R, Mandal M, et al. Transcriptional repression of oestrogen receptor by metastasis-associated protein 1 corepressor. Nat Cell Biol. 2001;3(1):30–7.CrossRefPubMed Mazumdar A, Wang RA, Mishra SK, Adam L, Bagheri-Yarmand R, Mandal M, et al. Transcriptional repression of oestrogen receptor by metastasis-associated protein 1 corepressor. Nat Cell Biol. 2001;3(1):30–7.CrossRefPubMed
34.
go back to reference Brehm A, Miska EA, McCance DJ, Reid JL, Bannister AJ, Kouzarides T. Retinoblastoma protein recruits histone deacetylase to repress transcription. Nature. 1998;391(6667):597–601.CrossRefPubMed Brehm A, Miska EA, McCance DJ, Reid JL, Bannister AJ, Kouzarides T. Retinoblastoma protein recruits histone deacetylase to repress transcription. Nature. 1998;391(6667):597–601.CrossRefPubMed
35.
go back to reference Magnaghi-Jaulin L, Groisman R, Naguibneva I, Robin P, Lorain S, Le Villain JP, et al. Retinoblastoma protein represses transcription by recruiting a histone deacetylase. Nature. 1998;391(6667):601–5.CrossRefPubMed Magnaghi-Jaulin L, Groisman R, Naguibneva I, Robin P, Lorain S, Le Villain JP, et al. Retinoblastoma protein represses transcription by recruiting a histone deacetylase. Nature. 1998;391(6667):601–5.CrossRefPubMed
36.
go back to reference Luo RX, Postigo AA, Dean DC. Rb interacts with histone deacetylase to repress transcription. Cell. 1998;92(4):463–73.CrossRefPubMed Luo RX, Postigo AA, Dean DC. Rb interacts with histone deacetylase to repress transcription. Cell. 1998;92(4):463–73.CrossRefPubMed
37.
go back to reference Jaejoon Won JY, Tae Kook K. Sp1 and Sp3 recruit histone deacetylase to repress transcription of human telomerase reverse transcriptase (hTERT) promoter in normal human somatic cells. J Biol Chem. 2002;277:38230–8.CrossRefPubMed Jaejoon Won JY, Tae Kook K. Sp1 and Sp3 recruit histone deacetylase to repress transcription of human telomerase reverse transcriptase (hTERT) promoter in normal human somatic cells. J Biol Chem. 2002;277:38230–8.CrossRefPubMed
38.
go back to reference Chang HC, Cho CY, Hung WC. Downregulation of RECK by promoter methylation correlates with lymph node metastasis in non-small cell lung cancer. Cancer Sci. 2007;98(2):169–73.CrossRefPubMed Chang HC, Cho CY, Hung WC. Downregulation of RECK by promoter methylation correlates with lymph node metastasis in non-small cell lung cancer. Cancer Sci. 2007;98(2):169–73.CrossRefPubMed
39.
go back to reference Webb CP, Van Aelst L, Wigler MH, Woude GF. Signaling pathways in Ras-mediated tumorigenicity and metastasis. Proc Natl Acad Sci U S A. 1998;95(15):8773–8.CrossRefPubMedPubMedCentral Webb CP, Van Aelst L, Wigler MH, Woude GF. Signaling pathways in Ras-mediated tumorigenicity and metastasis. Proc Natl Acad Sci U S A. 1998;95(15):8773–8.CrossRefPubMedPubMedCentral
40.
go back to reference Welch DR, Sakamaki T, Pioquinto R, Leonard TO, Goldberg SF, Hon Q, et al. Transfection of constitutively active mitogen-activated protein/extracellular signal-regulated kinase kinase confers tumorigenic and metastatic potentials to NIH3T3 cells. Cancer Res. 2000;60(6):1552–6.PubMed Welch DR, Sakamaki T, Pioquinto R, Leonard TO, Goldberg SF, Hon Q, et al. Transfection of constitutively active mitogen-activated protein/extracellular signal-regulated kinase kinase confers tumorigenic and metastatic potentials to NIH3T3 cells. Cancer Res. 2000;60(6):1552–6.PubMed
41.
go back to reference Guan LS, Rauchman M, Wang ZY. Induction of Rb-associated protein (RbAp46) by Wilms’ tumor suppressor WT1 mediates growth inhibition. J Biol Chem. 1998;273(42):27047–50.CrossRefPubMed Guan LS, Rauchman M, Wang ZY. Induction of Rb-associated protein (RbAp46) by Wilms’ tumor suppressor WT1 mediates growth inhibition. J Biol Chem. 1998;273(42):27047–50.CrossRefPubMed
42.
go back to reference Yao YL, Yang WM. The metastasis-associated proteins 1 and 2 form distinct protein complexes with histone deacetylase activity. J Biol Chem. 2003;278(43):42560–8.CrossRefPubMed Yao YL, Yang WM. The metastasis-associated proteins 1 and 2 form distinct protein complexes with histone deacetylase activity. J Biol Chem. 2003;278(43):42560–8.CrossRefPubMed
43.
go back to reference Toh Y, Kuninaka S, Endo K, Oshiro T, Ikeda Y, Nakashima H, et al. Molecular analysis of a candidate metastasis-associated gene, MTA1: possible interaction with histone deacetylase 1. J Exp Clin Cancer Res. 2000;19(1):105–11.PubMed Toh Y, Kuninaka S, Endo K, Oshiro T, Ikeda Y, Nakashima H, et al. Molecular analysis of a candidate metastasis-associated gene, MTA1: possible interaction with histone deacetylase 1. J Exp Clin Cancer Res. 2000;19(1):105–11.PubMed
44.
go back to reference Toh Y, Oki E, Oda S, Tokunaga E, Ohno S, Maehara Y, et al. Overexpression of the MTA1 gene in gastrointestinal carcinomas: correlation with invasion and metastasis. Int J Cancer. 1997;74(4):459–63.CrossRefPubMed Toh Y, Oki E, Oda S, Tokunaga E, Ohno S, Maehara Y, et al. Overexpression of the MTA1 gene in gastrointestinal carcinomas: correlation with invasion and metastasis. Int J Cancer. 1997;74(4):459–63.CrossRefPubMed
45.
go back to reference Toh Y, Kuwano H, Mori M, Nicolson GL, Sugimachi K. Overexpression of metastasis-associated MTA1 mRNA in invasive oesophageal carcinomas. Br J Cancer. 1999;79(11–12):1723–6.CrossRefPubMedPubMedCentral Toh Y, Kuwano H, Mori M, Nicolson GL, Sugimachi K. Overexpression of metastasis-associated MTA1 mRNA in invasive oesophageal carcinomas. Br J Cancer. 1999;79(11–12):1723–6.CrossRefPubMedPubMedCentral
46.
go back to reference Nicolas E, Morales V, Magnaghi-Jaulin L, Harel-Bellan A, Richard-Foy H, Trouche D. RbAp48 belongs to the histone deacetylase complex that associates with the retinoblastoma protein. J Biol Chem. 2000;275(13):9797–804.CrossRefPubMed Nicolas E, Morales V, Magnaghi-Jaulin L, Harel-Bellan A, Richard-Foy H, Trouche D. RbAp48 belongs to the histone deacetylase complex that associates with the retinoblastoma protein. J Biol Chem. 2000;275(13):9797–804.CrossRefPubMed
Metadata
Title
Ras induces experimental lung metastasis through up-regulation of RbAp46 to suppress RECK promoter activity
Authors
Hsuan-Heng Yeh
Yu-Fen Tseng
Yu-Chiao Hsu
Sheng-Hui Lan
Shan-Ying Wu
Giri Raghavaraju
Da-En Cheng
Ying-Ray Lee
Tsuey-Yu Chang
Nan-Haw Chow
Wen-Chun Hung
Hsiao-Sheng Liu
Publication date
01-12-2015
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2015
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-015-1155-7

Other articles of this Issue 1/2015

BMC Cancer 1/2015 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