Skip to main content
Top
Published in: BMC Cancer 1/2018

Open Access 01-12-2018 | Research article

Histone methyltransferase SETDB1 promotes cells proliferation and migration by interacting withTiam1 in hepatocellular carcinoma

Authors: Yuqin Zhang, Jing Huang, Qisheng Li, Keli Chen, Yonghao Liang, Zetao Zhan, Feng Ye, Wen Ni, Longhua Chen, Yi Ding

Published in: BMC Cancer | Issue 1/2018

Login to get access

Abstract

Background

SETDB1 is a histone H3K9 methyltransferase, which plays a significant role in the occurrence and progression of tumors. Previous studies have confirmed that T-lymphom invasion and metastasis gene (Tiam1) is a protein associated with the metastasis of hepatocellular carcinoma (HCC); however, we have not yet been successful in elucidating the specific mechanism of HCC.

Methods

Yeast two-hybrid test was conducted to screen proteins that interacted with Tiam1 gene. Glutathione-S-transferase (GST) pull-down and crosslinking-immunoprecipitation (CLIP) assays were performed to determine whether SETDB1 can interact with Tiam1 gene. A series of related experiments were performed to explore role of SETDB1 on cell proliferation, migration, and invasion in HCC. Recovery experiment was performed to investigate the effect of Tiam1 knockdown on cell proliferation and migration, which was caused by SETDB1 overexpression in HCC cells. The expression of SETDB1 was frequently upregulated in HCC tissues and positively correlated with Tiam1.

Results

GST pull-down and CLIP assays were performed to elucidate the interaction between SETDB1 and Tiam1. Cell proliferation, migration, and epithelial mesenchymal transformation (EMT) in HCC cells was promoted with the overexpression of SETDB1. Following the knockdown of Tiam1 gene, the effect of SETDB1 on cell proliferation and migration was reversed in HCC cells. The expression of SETDB1 was frequently up-regulated in HCC tissues, and it was positively correlated with Tiam1 gene.

Conclusions

Ours is the first study to prove that SETDB1 promotes the proliferation and migration of cells by forming SETDB1-Tiam1 compounds. We found that SETDB1-Tiam1 compounds were involved in a novel pathway, which regulated epigenetic modification of gene expression in HCC samples.
Appendix
Available only for authorised users
Literature
2.
go back to reference Budhu A, et al. Prediction of venous metastases, recurrence, and prognosis in hepatocellular carcinoma based on a unique immune response signature of the liver microenvironment. Cancer Cell. 2006;10(2):99–111.CrossRefPubMed Budhu A, et al. Prediction of venous metastases, recurrence, and prognosis in hepatocellular carcinoma based on a unique immune response signature of the liver microenvironment. Cancer Cell. 2006;10(2):99–111.CrossRefPubMed
3.
go back to reference Fidler IJ. The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis revisited. Nat Rev Cancer. 2003;3(6):453–8.CrossRefPubMed Fidler IJ. The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis revisited. Nat Rev Cancer. 2003;3(6):453–8.CrossRefPubMed
4.
go back to reference Huang J, et al. Tiam1 is associated with hepatocellular carcinoma metastasis. Int J Cancer. 2013;132(1):90–100.CrossRefPubMed Huang J, et al. Tiam1 is associated with hepatocellular carcinoma metastasis. Int J Cancer. 2013;132(1):90–100.CrossRefPubMed
5.
go back to reference Shinjo K, Kondo Y. Clinical implications of epigenetic alterations in human thoracic malignancies: epigenetic alterations in lung cancer. Methods Mol Biol. 2012;863:221–39.CrossRefPubMed Shinjo K, Kondo Y. Clinical implications of epigenetic alterations in human thoracic malignancies: epigenetic alterations in lung cancer. Methods Mol Biol. 2012;863:221–39.CrossRefPubMed
6.
go back to reference Ellinger J, et al. Global levels of histone modifications predict prostate cancer recurrence. Prostate. 2010;70(1):61–9.CrossRefPubMed Ellinger J, et al. Global levels of histone modifications predict prostate cancer recurrence. Prostate. 2010;70(1):61–9.CrossRefPubMed
8.
go back to reference Park YS, et al. The global histone modification pattern correlates with cancer recurrence and overall survival in gastric adenocarcinoma. Ann Surg Oncol. 2008;15(7):1968–76.CrossRefPubMed Park YS, et al. The global histone modification pattern correlates with cancer recurrence and overall survival in gastric adenocarcinoma. Ann Surg Oncol. 2008;15(7):1968–76.CrossRefPubMed
9.
go back to reference Song JS, et al. Global histone modification pattern associated with recurrence and disease-free survival in non-small cell lung cancer patients. Pathol Int. 2012;62(3):182–90.CrossRefPubMed Song JS, et al. Global histone modification pattern associated with recurrence and disease-free survival in non-small cell lung cancer patients. Pathol Int. 2012;62(3):182–90.CrossRefPubMed
10.
go back to reference Schultz DC, et al. SETDB1: a novel KAP-1-associated histone H3, lysine 9-specific methyltransferase that contributes to HP1-mediated silencing of euchromatic genes by KRAB zinc-finger proteins. Genes Dev. 2002;16(8):919–32.CrossRefPubMedPubMedCentral Schultz DC, et al. SETDB1: a novel KAP-1-associated histone H3, lysine 9-specific methyltransferase that contributes to HP1-mediated silencing of euchromatic genes by KRAB zinc-finger proteins. Genes Dev. 2002;16(8):919–32.CrossRefPubMedPubMedCentral
11.
go back to reference Chen MW, et al. H3K9 histone methyltransferase G9a promotes lung cancer invasion and metastasis by silencing the cell adhesion molecule ep-CAM. Cancer Res. 2010;70(20):7830–40.CrossRefPubMed Chen MW, et al. H3K9 histone methyltransferase G9a promotes lung cancer invasion and metastasis by silencing the cell adhesion molecule ep-CAM. Cancer Res. 2010;70(20):7830–40.CrossRefPubMed
12.
go back to reference Kondo Y, et al. Downregulation of histone H3 lysine 9 methyltransferase G9a induces centrosome disruption and chromosome instability in cancer cells. PLoS One. 2008;3(4):e2037.CrossRefPubMedPubMedCentral Kondo Y, et al. Downregulation of histone H3 lysine 9 methyltransferase G9a induces centrosome disruption and chromosome instability in cancer cells. PLoS One. 2008;3(4):e2037.CrossRefPubMedPubMedCentral
13.
go back to reference Watanabe H, et al. Deregulation of histone lysine methyltransferases contributes to oncogenic transformation of human bronchoepithelial cells. Cancer Cell Int. 2008;8:15.CrossRefPubMedPubMedCentral Watanabe H, et al. Deregulation of histone lysine methyltransferases contributes to oncogenic transformation of human bronchoepithelial cells. Cancer Cell Int. 2008;8:15.CrossRefPubMedPubMedCentral
14.
go back to reference Zhang Y, et al. MiR-20a induces cell Radioresistance by activating the PTEN/PI3K/Akt signaling pathway in hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2015;92(5):1132–40.CrossRefPubMed Zhang Y, et al. MiR-20a induces cell Radioresistance by activating the PTEN/PI3K/Akt signaling pathway in hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2015;92(5):1132–40.CrossRefPubMed
15.
go back to reference Chen JS, et al. Expression of T-cell lymphoma invasion and metastasis 2 (TIAM2) promotes proliferation and invasion of liver cancer. Int J Cancer. 2012;130(6):1302–13.CrossRefPubMed Chen JS, et al. Expression of T-cell lymphoma invasion and metastasis 2 (TIAM2) promotes proliferation and invasion of liver cancer. Int J Cancer. 2012;130(6):1302–13.CrossRefPubMed
16.
go back to reference Zhu JM, Yu PW. Downregulation of Tcell lymphoma invasion and metastasisinducing factor 1 induces cytoskeletal rearrangement and inhibits the invasive capacity of gastric cancer cells. Mol Med Rep. 2013;8(2):425–33.CrossRefPubMed Zhu JM, Yu PW. Downregulation of Tcell lymphoma invasion and metastasisinducing factor 1 induces cytoskeletal rearrangement and inhibits the invasive capacity of gastric cancer cells. Mol Med Rep. 2013;8(2):425–33.CrossRefPubMed
17.
18.
go back to reference Zhao L, et al. Overexpression of T lymphoma invasion and metastasis 1 predict renal cell carcinoma metastasis and overall patient survival. J Cancer Res Clin Oncol. 2011;137(3):393–8.CrossRefPubMed Zhao L, et al. Overexpression of T lymphoma invasion and metastasis 1 predict renal cell carcinoma metastasis and overall patient survival. J Cancer Res Clin Oncol. 2011;137(3):393–8.CrossRefPubMed
19.
go back to reference Hu J, et al. The downregulation of MiR-182 is associated with the growth and invasion of osteosarcoma cells through the regulation of TIAM1 expression. PLoS One. 2015;10(5):e0121175.CrossRefPubMedPubMedCentral Hu J, et al. The downregulation of MiR-182 is associated with the growth and invasion of osteosarcoma cells through the regulation of TIAM1 expression. PLoS One. 2015;10(5):e0121175.CrossRefPubMedPubMedCentral
20.
go back to reference Li Z, et al. By downregulating TIAM1 expression, microRNA-329 suppresses gastric cancer invasion and growth. Oncotarget. 2015;6(19):17559–69.PubMedPubMedCentral Li Z, et al. By downregulating TIAM1 expression, microRNA-329 suppresses gastric cancer invasion and growth. Oncotarget. 2015;6(19):17559–69.PubMedPubMedCentral
21.
go back to reference Whalley HJ, et al. Cdk1 phosphorylates the Rac activator Tiam1 to activate centrosomal Pak and promote mitotic spindle formation. Nat Commun. 2015;6:7437.CrossRefPubMedPubMedCentral Whalley HJ, et al. Cdk1 phosphorylates the Rac activator Tiam1 to activate centrosomal Pak and promote mitotic spindle formation. Nat Commun. 2015;6:7437.CrossRefPubMedPubMedCentral
22.
go back to reference Wang S, et al. Tiam1 interaction with the PAR complex promotes Talin-mediated Rac1 activation during polarized cell migration. J Cell Biol. 2012;199(2):331–45.CrossRefPubMedPubMedCentral Wang S, et al. Tiam1 interaction with the PAR complex promotes Talin-mediated Rac1 activation during polarized cell migration. J Cell Biol. 2012;199(2):331–45.CrossRefPubMedPubMedCentral
23.
go back to reference Rajagopal S, et al. Scaffold proteins IRSp53 and spinophilin regulate localized Rac activation by T-lymphocyte invasion and metastasis protein 1 (TIAM1). J Biol Chem. 2010;285(23):18060–71.CrossRefPubMedPubMedCentral Rajagopal S, et al. Scaffold proteins IRSp53 and spinophilin regulate localized Rac activation by T-lymphocyte invasion and metastasis protein 1 (TIAM1). J Biol Chem. 2010;285(23):18060–71.CrossRefPubMedPubMedCentral
25.
26.
go back to reference Bilodeau S, et al. SetDB1 contributes to repression of genes encoding developmental regulators and maintenance of ES cell state. Genes Dev. 2009;23(21):2484–9.CrossRefPubMedPubMedCentral Bilodeau S, et al. SetDB1 contributes to repression of genes encoding developmental regulators and maintenance of ES cell state. Genes Dev. 2009;23(21):2484–9.CrossRefPubMedPubMedCentral
27.
go back to reference Yuan P, et al. Eset partners with Oct4 to restrict extraembryonic trophoblast lineage potential in embryonic stem cells. Genes Dev. 2009;23(21):2507–20.CrossRefPubMedPubMedCentral Yuan P, et al. Eset partners with Oct4 to restrict extraembryonic trophoblast lineage potential in embryonic stem cells. Genes Dev. 2009;23(21):2507–20.CrossRefPubMedPubMedCentral
28.
go back to reference Rodriguez-Paredes M, et al. Gene amplification of the histone methyltransferase SETDB1 contributes to human lung tumorigenesis. Oncogene. 2014;33(21):2807–13.CrossRefPubMed Rodriguez-Paredes M, et al. Gene amplification of the histone methyltransferase SETDB1 contributes to human lung tumorigenesis. Oncogene. 2014;33(21):2807–13.CrossRefPubMed
29.
go back to reference Sun QY, et al. SETDB1 accelerates tumourigenesis by regulating the WNT signalling pathway. J Pathol. 2015;235(4):559–70.CrossRefPubMed Sun QY, et al. SETDB1 accelerates tumourigenesis by regulating the WNT signalling pathway. J Pathol. 2015;235(4):559–70.CrossRefPubMed
30.
Metadata
Title
Histone methyltransferase SETDB1 promotes cells proliferation and migration by interacting withTiam1 in hepatocellular carcinoma
Authors
Yuqin Zhang
Jing Huang
Qisheng Li
Keli Chen
Yonghao Liang
Zetao Zhan
Feng Ye
Wen Ni
Longhua Chen
Yi Ding
Publication date
01-12-2018
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2018
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-4464-9

Other articles of this Issue 1/2018

BMC Cancer 1/2018 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