Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2019

Open Access 01-12-2019 | Metastasis | Research

The RNA binding protein RBMS3 inhibits the metastasis of breast cancer by regulating Twist1 expression

Authors: Lei Zhu, Pei-Wen Xi, Xiao-Xia Li, Xi Sun, Wen-Bin Zhou, Tian-Song Xia, Liang Shi, Yue Hu, Qiang Ding, Ji-Fu Wei

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2019

Login to get access

Abstract

Background

Metastasis remains the biggest obstacle for breast cancer treatment. Therefore, identification of specific biomarker of metastasis is very necessary. The RNA binding protein 3 (RBMS3) acts as a tumor suppressor in various cancers. Whereas, its role and underlying molecular mechanism in breast cancer is far from elucidated.

Methods

Quantitative real-time PCR and western blots were carried out to determine the expression of RBMS3 in breast cancer cells and tissues. Transwell and in vivo metastasis assay were conducted to investigate the effects of RBMS3 on migration, invasion and metastasis of breast cancer cells. Transcriptome sequencing was applied to screen out the differential gene expression affected by RBMS3. RNA immunoprecipitation assay combined with luciferase reporter assay were performed to explore the direct correlation between RBMS3 and Twist1 mRNA.

Results

RBMS3 was downregulated in breast cancer and ectopic expression of RBMS3 contributed to inhibition of cell migration, invasion in vitro and lung metastasis in vivo. Furthermore, RBMS3 negatively regulated Twsit1 expression via directly binding to 3′-UTR of Twist1 mRNA, and thereby decreased Twist1-induced expression of matrix metalloproteinase 2 (MMP2). Additionally, Twist1-induced cell migration, invasion and lung metastasis could be reversed by the upregulation of RBMS3.

Conclusions

In summary, our study revealed a novel mechanism of the RBMS3/Twsit1/MMP2 axis in the regulation of invasion and metastasis of breast cancer, which may become a potential molecular marker for breast cancer treatment.
Literature
1.
go back to reference Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin [Journal Article]. 2015;65:87–108.CrossRef Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin [Journal Article]. 2015;65:87–108.CrossRef
2.
go back to reference Ufen MP, Köhne CH, Wischneswky M, Wolters R, Novopashenny I, Fischer J, et al. Metastatic breast cancer: are we treating the same patients as in the past? Ann Oncol. 2014;25:95–100.CrossRef Ufen MP, Köhne CH, Wischneswky M, Wolters R, Novopashenny I, Fischer J, et al. Metastatic breast cancer: are we treating the same patients as in the past? Ann Oncol. 2014;25:95–100.CrossRef
3.
go back to reference Gupta GP, Massagué J. Cancer metastasis: building a framework. Cell. 2006;127:679–95.CrossRef Gupta GP, Massagué J. Cancer metastasis: building a framework. Cell. 2006;127:679–95.CrossRef
4.
go back to reference Fidler IJ. The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis revisited. Nat Rev Cancer. 2003;3:453.CrossRef Fidler IJ. The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis revisited. Nat Rev Cancer. 2003;3:453.CrossRef
5.
go back to reference Thiery JP, Acloque H, Huang RYJ, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–90.CrossRef Thiery JP, Acloque H, Huang RYJ, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–90.CrossRef
6.
go back to reference Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19:1423–37.CrossRef Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19:1423–37.CrossRef
7.
go back to reference Nguyen DX, Chiang AC, Zhang XHF, Kim JY, Kris MG, Ladanyi M, et al. WNT/TCF signaling through LEF1 and HOXB9 mediates lung adenocarcinoma metastasis. Cell. 2009;138:51–62.CrossRef Nguyen DX, Chiang AC, Zhang XHF, Kim JY, Kris MG, Ladanyi M, et al. WNT/TCF signaling through LEF1 and HOXB9 mediates lung adenocarcinoma metastasis. Cell. 2009;138:51–62.CrossRef
8.
go back to reference Giampieri S, Manning C, Hooper S, Jones L, Hill CS, Sahai E. Localised and reversible TGFβ signalling switches breast cancer cells from cohesive to single cell motility. Nat Cell Biol. 2009;11:1287–96.CrossRef Giampieri S, Manning C, Hooper S, Jones L, Hill CS, Sahai E. Localised and reversible TGFβ signalling switches breast cancer cells from cohesive to single cell motility. Nat Cell Biol. 2009;11:1287–96.CrossRef
9.
go back to reference Sullivan NJ, Sasser AK, Axel AE, Vesuna F, Raman V, Ramirez N, et al. Interleukin-6 induces an epithelial–mesenchymal transition phenotype in human breast cancer cells. Oncogene. 2009;28:2940–7.CrossRef Sullivan NJ, Sasser AK, Axel AE, Vesuna F, Raman V, Ramirez N, et al. Interleukin-6 induces an epithelial–mesenchymal transition phenotype in human breast cancer cells. Oncogene. 2009;28:2940–7.CrossRef
10.
go back to reference Clapéron A, Mergey M, Nguyen Ho-Bouldoires TH, Vignjevic D, Wendum D, Chrétien Y, et al. EGF/EGFR axis contributes to the progression of cholangiocarcinoma through the induction of an epithelial-mesenchymal transition. J Hepatol. 2014;61:325–32.CrossRef Clapéron A, Mergey M, Nguyen Ho-Bouldoires TH, Vignjevic D, Wendum D, Chrétien Y, et al. EGF/EGFR axis contributes to the progression of cholangiocarcinoma through the induction of an epithelial-mesenchymal transition. J Hepatol. 2014;61:325–32.CrossRef
11.
go back to reference Valastyan S, Weinberg RA. Tumor metastasis: molecular insights and evolving paradigms. Cell. 2011;147:275–92.CrossRef Valastyan S, Weinberg RA. Tumor metastasis: molecular insights and evolving paradigms. Cell. 2011;147:275–92.CrossRef
12.
go back to reference Wan L, Pantel K, Kang Y. Tumor metastasis: moving new biological insights into the clinic. Nat Med. 2013;19:1450.CrossRef Wan L, Pantel K, Kang Y. Tumor metastasis: moving new biological insights into the clinic. Nat Med. 2013;19:1450.CrossRef
13.
go back to reference Gupta GP, Nguyen DX, Chiang AC, Bos PD, Kim JY, Nadal C, et al. Mediators of vascular remodelling co-opted for sequential steps in lung metastasis. Nature. 2007;446:765.CrossRef Gupta GP, Nguyen DX, Chiang AC, Bos PD, Kim JY, Nadal C, et al. Mediators of vascular remodelling co-opted for sequential steps in lung metastasis. Nature. 2007;446:765.CrossRef
14.
go back to reference Wu J, Zhou X, Sun X, Xia T, Li X, Shi L, et al. RBM38 is involved in TGF-β-induced epithelial-to-mesenchymal transition by stabilising zonula occludens-1 mRNA in breast cancer. Brit J Cancer. 2017;117:675.CrossRef Wu J, Zhou X, Sun X, Xia T, Li X, Shi L, et al. RBM38 is involved in TGF-β-induced epithelial-to-mesenchymal transition by stabilising zonula occludens-1 mRNA in breast cancer. Brit J Cancer. 2017;117:675.CrossRef
15.
go back to reference Vanharanta S, Marney CB, Shu W, Valiente M, Zou Y, Mele A, et al. Loss of the multifunctional RNA-binding protein RBM47 as a source of selectable metastatic traits in breast cancer. Elife. 2014;e2734:3. Vanharanta S, Marney CB, Shu W, Valiente M, Zou Y, Mele A, et al. Loss of the multifunctional RNA-binding protein RBM47 as a source of selectable metastatic traits in breast cancer. Elife. 2014;e2734:3.
16.
go back to reference Gerstberger S, Hafner M, Tuschl T. A census of human RNA-binding proteins. Nat Rev Genet. 2014;15:829.CrossRef Gerstberger S, Hafner M, Tuschl T. A census of human RNA-binding proteins. Nat Rev Genet. 2014;15:829.CrossRef
17.
go back to reference Penkov D, Ni R, Else C, Piñol-Roma S, Ramirez F, Tanaka S. Cloning of a human gene closely related to the genes coding for the c-myc single-strand binding proteins. Gene. 2000;243:27–36.CrossRef Penkov D, Ni R, Else C, Piñol-Roma S, Ramirez F, Tanaka S. Cloning of a human gene closely related to the genes coding for the c-myc single-strand binding proteins. Gene. 2000;243:27–36.CrossRef
18.
go back to reference Li Y, Chen L, Nie C, Zeng T, Liu H, Mao X, et al. Downregulation of RBMS3 is associated with poor prognosis in esophageal squamous cell carcinoma. Cancer Res. 2011;71:6106.CrossRef Li Y, Chen L, Nie C, Zeng T, Liu H, Mao X, et al. Downregulation of RBMS3 is associated with poor prognosis in esophageal squamous cell carcinoma. Cancer Res. 2011;71:6106.CrossRef
19.
go back to reference Liang Y, Liu Y, Meng Q, Li X, Wang F, Yao G, et al. RBMS3 is a tumor suppressor gene that acts as a favorable prognostic marker in lung squamous cell carcinoma. Med Oncol. 2015;32:30.CrossRef Liang Y, Liu Y, Meng Q, Li X, Wang F, Yao G, et al. RBMS3 is a tumor suppressor gene that acts as a favorable prognostic marker in lung squamous cell carcinoma. Med Oncol. 2015;32:30.CrossRef
20.
go back to reference Chen J, Kwong DL, Zhu C, Chen L, Dong S, Zhang L, et al. RBMS3 at 3p24 inhibits nasopharyngeal carcinoma development via inhibiting cell proliferation, angiogenesis, and Inducing Apoptosis. Plos One. 2012;7:e44636.CrossRef Chen J, Kwong DL, Zhu C, Chen L, Dong S, Zhang L, et al. RBMS3 at 3p24 inhibits nasopharyngeal carcinoma development via inhibiting cell proliferation, angiogenesis, and Inducing Apoptosis. Plos One. 2012;7:e44636.CrossRef
21.
go back to reference Zhang T, Wu Y, Fang Z, Yan Q, Zhang S, Sun R, et al. Low expression of RBMS3 and SFRP1 are associated with poor prognosis in patients with gastric cancer. Am J Cancer Res. 2016;6:2679–89.PubMedPubMedCentral Zhang T, Wu Y, Fang Z, Yan Q, Zhang S, Sun R, et al. Low expression of RBMS3 and SFRP1 are associated with poor prognosis in patients with gastric cancer. Am J Cancer Res. 2016;6:2679–89.PubMedPubMedCentral
22.
go back to reference Wu G, Cao L, Zhu J, Tan Z, Tang M, Li Z, et al. Loss of RBMS3 confers platinum-resistance in epithelial ovarian Cancer via activation of miR-126-5p/β-catenin/CBP signaling. In: Clin Cancer res; 2018. Wu G, Cao L, Zhu J, Tan Z, Tang M, Li Z, et al. Loss of RBMS3 confers platinum-resistance in epithelial ovarian Cancer via activation of miR-126-5p/β-catenin/CBP signaling. In: Clin Cancer res; 2018.
23.
go back to reference Yang Y, Quan L, Ling Y. RBMS3 inhibits the proliferation and metastasis of breast Cancer cells. Oncol Res. 2018;26:9–15.CrossRef Yang Y, Quan L, Ling Y. RBMS3 inhibits the proliferation and metastasis of breast Cancer cells. Oncol Res. 2018;26:9–15.CrossRef
24.
go back to reference Rahme GJ, Israel MA. Id4 suppresses MMP2-mediated invasion of glioblastoma-derived cells by direct inactivation of Twist1 function. Oncogene. 2014;34:53.CrossRef Rahme GJ, Israel MA. Id4 suppresses MMP2-mediated invasion of glioblastoma-derived cells by direct inactivation of Twist1 function. Oncogene. 2014;34:53.CrossRef
25.
go back to reference Zhou X, Wu J, Shi L, Li X, Zhu L, Sun X, et al. PTEN expression is upregulated by a RNA-binding protein RBM38 via enhancing its mRNA stability in breast cancer. J Exp Clin Canc Res. 2017;36:149.CrossRef Zhou X, Wu J, Shi L, Li X, Zhu L, Sun X, et al. PTEN expression is upregulated by a RNA-binding protein RBM38 via enhancing its mRNA stability in breast cancer. J Exp Clin Canc Res. 2017;36:149.CrossRef
26.
go back to reference Shi L, Xia T, Wei X, Zhou W, Xue J, Cheng L, et al. Estrogen receptor (ER) was regulated by RNPC1 stabilizing mRNA in ER positive breast cancer. Oncotarget. 2015;6:12264.PubMedPubMedCentral Shi L, Xia T, Wei X, Zhou W, Xue J, Cheng L, et al. Estrogen receptor (ER) was regulated by RNPC1 stabilizing mRNA in ER positive breast cancer. Oncotarget. 2015;6:12264.PubMedPubMedCentral
27.
go back to reference Li X, Shi L, Zhou X, Wu J, Xia T, Zhou W, et al. The role of c-Myc-RBM38 loop in the growth suppression in breast cancer. J Exp Clin Canc Res. 2017;36:49.CrossRef Li X, Shi L, Zhou X, Wu J, Xia T, Zhou W, et al. The role of c-Myc-RBM38 loop in the growth suppression in breast cancer. J Exp Clin Canc Res. 2017;36:49.CrossRef
28.
go back to reference Jayasena CS, Bronner ME. Rbms3 functions in craniofacial development by posttranscriptionally modulating TGF-β signaling. J Cell Biol. 2012;199:453–66.CrossRef Jayasena CS, Bronner ME. Rbms3 functions in craniofacial development by posttranscriptionally modulating TGF-β signaling. J Cell Biol. 2012;199:453–66.CrossRef
29.
go back to reference Wu Y, Yun D, Zhao Y, Wang Y, Sun R, Yan Q, et al. Down regulation of RNA binding motif, single-stranded interacting protein 3, along with up regulation of nuclear HIF1A correlates with poor prognosis in patients with gastric cancer. Oncotarget. 2017;8:1262–77.PubMed Wu Y, Yun D, Zhao Y, Wang Y, Sun R, Yan Q, et al. Down regulation of RNA binding motif, single-stranded interacting protein 3, along with up regulation of nuclear HIF1A correlates with poor prognosis in patients with gastric cancer. Oncotarget. 2017;8:1262–77.PubMed
30.
go back to reference Fritz D, Stefanovic B. RNA binding protein RBMS3 is expressed in activated hepatic stellate cells and liver fibrosis and increases expression of transcription factor Prx1. J Mol Biol. 2007;371:585–95.CrossRef Fritz D, Stefanovic B. RNA binding protein RBMS3 is expressed in activated hepatic stellate cells and liver fibrosis and increases expression of transcription factor Prx1. J Mol Biol. 2007;371:585–95.CrossRef
31.
go back to reference Chang HY, Nuyten DSA, Sneddon JB, Hastie T, Tibshirani R, Sørlie T, et al. Robustness, scalability, and integration of a wound-response gene expression signature in predicting breast cancer survival. P Natl Acad Sci U S A. 2005;102:3738–43.CrossRef Chang HY, Nuyten DSA, Sneddon JB, Hastie T, Tibshirani R, Sørlie T, et al. Robustness, scalability, and integration of a wound-response gene expression signature in predicting breast cancer survival. P Natl Acad Sci U S A. 2005;102:3738–43.CrossRef
32.
go back to reference Gilkes DM, Semenza GL, Wirtz D. Hypoxia and the extracellular matrix: drivers of tumour metastasis. Nat Rev Cancer. 2014;14:430–9.CrossRef Gilkes DM, Semenza GL, Wirtz D. Hypoxia and the extracellular matrix: drivers of tumour metastasis. Nat Rev Cancer. 2014;14:430–9.CrossRef
33.
go back to reference Eckert MA, Lwin TM, Chang AT, Kim J, Danis E, Ohno-Machado L, et al. Twist1-induced invadopodia formation promotes tumor metastasis. Cancer Cell. 2011;19:372–86.CrossRef Eckert MA, Lwin TM, Chang AT, Kim J, Danis E, Ohno-Machado L, et al. Twist1-induced invadopodia formation promotes tumor metastasis. Cancer Cell. 2011;19:372–86.CrossRef
34.
go back to reference Lee K, Yeo S, Sung CO, Kim S. Twist1 is a key regulator of Cancer-associated fibroblasts. Cancer Res. 2015;75:73.CrossRef Lee K, Yeo S, Sung CO, Kim S. Twist1 is a key regulator of Cancer-associated fibroblasts. Cancer Res. 2015;75:73.CrossRef
35.
go back to reference Pickup MW, Mouw JK, Weaver VM. The extracellular matrix modulates the hallmarks of cancer. EMBO Rep. 2014;15:1243–53.CrossRef Pickup MW, Mouw JK, Weaver VM. The extracellular matrix modulates the hallmarks of cancer. EMBO Rep. 2014;15:1243–53.CrossRef
36.
go back to reference Slattery ML, John E, Torres-Mejia G, Stern M, Lundgreen A, Hines L, et al. Matrix metalloproteinase genes are associated with breast Cancer risk and survival: the breast Cancer health disparities study. PLoS One. 2013;8:e63165.CrossRef Slattery ML, John E, Torres-Mejia G, Stern M, Lundgreen A, Hines L, et al. Matrix metalloproteinase genes are associated with breast Cancer risk and survival: the breast Cancer health disparities study. PLoS One. 2013;8:e63165.CrossRef
Metadata
Title
The RNA binding protein RBMS3 inhibits the metastasis of breast cancer by regulating Twist1 expression
Authors
Lei Zhu
Pei-Wen Xi
Xiao-Xia Li
Xi Sun
Wen-Bin Zhou
Tian-Song Xia
Liang Shi
Yue Hu
Qiang Ding
Ji-Fu Wei
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2019
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-019-1111-5

Other articles of this Issue 1/2019

Journal of Experimental & Clinical Cancer Research 1/2019 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