Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01-12-2018 | Research

RBM38 plays a tumor-suppressor role via stabilizing the p53-mdm2 loop function in hepatocellular carcinoma

Authors: Jiazhou Ye, Rong Liang, Tao Bai, Yan Lin, Rongyun Mai, Meng Wei, Xinqin Ye, Lequn Li, Feixiang Wu

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2018

Login to get access

Abstract

Background

Misregulation of the p53-mdm2 loop function is a major mechanism to promote hepatocellular carcinoma (HCC). RBM38, a member of the RNA recognition motif (RRM) family of RNA binding proteins (RBPs), plays a fundamental role in the posttranscriptional control of gene expression and regulatory functions in human tumors. A novel RBM38-p53-mdm2 autoregulatory feedback loop has been demonstrated. However, its mechanistic role in HCC remains unclear.

Methods

In the present study, we investigated the role and molecular mechanism of misregulation in the p53-mdm2 loop function by RBM38 in HCC. First we investigated the correlation of RBM38 activity and p53-mdm2 loop function in liver cancer cells and HCC tissues by western blot and quantitative RT-PCR. We then conducted functional assays to investigate the molecular roles of RBM38 in inhibiting liver cancer cells aggressiveness in vitro and suppressing tumorigenicity in vivo.

Results

We observed RBM38 protein expression was commonly silenced coupled with increased mdm2 and decreased wild type (wt) p53 in liver cancer cells and HCC tissues compared to the corresponding normal liver cells and adjacent liver tissues. RBM38 mRNA level was significantly lower in HCC than adjacent liver tissues, whereas mdm2 and wtp53 mRNA levels were similar between HCC and adjacent liver tissues. This implied that deactivation of RBM38 could disrupt the p53-mdm2 loop and promote HCC, even though p53 and mdm2 transcript amounts were stable. Then, we generated stable liver cancer cell lines with overexpressed RBM38 (RBM38-OE) and found that up-regulation of RBM38 could inhibit mdm2 and restore wtp53 expression. Luciferase assay shown that RBM38 destabilized the mdm2 transcript through binding to multiple AU-/U-rich elements in mdm2 3’-UTR. Furthermore, functional assays showed that ectopic expression of RBM38 could induce liver cancer cell apoptosis and senescence, inhibit proliferation and colony growth, and suppress migration and invasion in vitro. Lastly, RBM38 could suppress HCC tumorigenicity in vivo.

Conclusion

Our findings suggested that RBM38 may be a core contributor in stabilizing the p53-mdm2 loop function to prevent HCC, and a potential novel target to provide a therapeutic strategy for HCC by inhibiting mdm2 and rescuing p53 from inactivation.
Appendix
Available only for authorised users
Literature
1.
go back to reference Torre LA, Bray F, Siegel RL, et al. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108. Torre LA, Bray F, Siegel RL, et al. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108.
2.
go back to reference Boyault S, Rickman DS, de Reynies A, et al. Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets. Hepatology. 2007;45:42–52.CrossRefPubMed Boyault S, Rickman DS, de Reynies A, et al. Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets. Hepatology. 2007;45:42–52.CrossRefPubMed
3.
go back to reference Pogribny IP, Rusyn I. Role of epigenetic aberrations in the development and progression of human hepatocellular carcinoma. Cancer Lett. 2014;342:223–30.CrossRefPubMed Pogribny IP, Rusyn I. Role of epigenetic aberrations in the development and progression of human hepatocellular carcinoma. Cancer Lett. 2014;342:223–30.CrossRefPubMed
5.
go back to reference Xue W, Zender L, Miething C, et al. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature. 2007;445:656–60.CrossRefPubMedPubMedCentral Xue W, Zender L, Miething C, et al. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature. 2007;445:656–60.CrossRefPubMedPubMedCentral
7.
go back to reference Momand J, Zambetti GP, Olson DC, et al. The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell. 1992;69:1237–45.CrossRefPubMed Momand J, Zambetti GP, Olson DC, et al. The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell. 1992;69:1237–45.CrossRefPubMed
8.
go back to reference Oliner JD, Pietenpol JA, Thiagalingam S, et al. Oncoprotein MDM2 conceals the activation domain of tumour suppressor p53. Nature. 1993;362:857–60.CrossRefPubMed Oliner JD, Pietenpol JA, Thiagalingam S, et al. Oncoprotein MDM2 conceals the activation domain of tumour suppressor p53. Nature. 1993;362:857–60.CrossRefPubMed
10.
go back to reference Zauberman A, Flusberg D, Haupt Y, et al. A functional p53-responsive intronic promoter is contained within the human mdm2 gene. Nucleic Acids Res. 1995;23:2584–92.CrossRefPubMedPubMedCentral Zauberman A, Flusberg D, Haupt Y, et al. A functional p53-responsive intronic promoter is contained within the human mdm2 gene. Nucleic Acids Res. 1995;23:2584–92.CrossRefPubMedPubMedCentral
11.
go back to reference Nishida N, Kudo M. Recent advancements in comprehensive genetic analyses for human hepatocellular carcinoma. Oncology. 2013;84(1):93–7.CrossRefPubMed Nishida N, Kudo M. Recent advancements in comprehensive genetic analyses for human hepatocellular carcinoma. Oncology. 2013;84(1):93–7.CrossRefPubMed
12.
go back to reference Nose H, Imazeki F, Ohto M, et al. p53 gene mutations and 17p allelic deletions in hepatocellular carcinoma from Japan. Cancer. 1993;72:355–60.CrossRefPubMed Nose H, Imazeki F, Ohto M, et al. p53 gene mutations and 17p allelic deletions in hepatocellular carcinoma from Japan. Cancer. 1993;72:355–60.CrossRefPubMed
13.
go back to reference Hussain SP, Schwank J, Staib F, et al. TP53 mutations and hepatocellular carcinoma: insights into the etiology and pathogenesis of liver cancer. Oncogene. 2007;26:2166–76.CrossRefPubMed Hussain SP, Schwank J, Staib F, et al. TP53 mutations and hepatocellular carcinoma: insights into the etiology and pathogenesis of liver cancer. Oncogene. 2007;26:2166–76.CrossRefPubMed
14.
go back to reference Meng X, Franklin DA, Dong J, et al. MDM2–p53 Pathway in Hepatocellular Carcinoma. Cancer Res. 2014;74(24):7161–7. Meng X, Franklin DA, Dong J, et al. MDM2–p53 Pathway in Hepatocellular Carcinoma. Cancer Res. 2014;74(24):7161–7.
16.
go back to reference Dharel N, Kato N, Muroyama R, et al. MDM2 promoter SNP309 is associated with the risk of hepatocellular carcinoma in patients with chronic hepatitis C. Clin Cancer Res. 2006;12:4867–71.CrossRefPubMed Dharel N, Kato N, Muroyama R, et al. MDM2 promoter SNP309 is associated with the risk of hepatocellular carcinoma in patients with chronic hepatitis C. Clin Cancer Res. 2006;12:4867–71.CrossRefPubMed
17.
go back to reference Yoon YJ, Chang HY, Ahn SH, et al. MDM2 and p53 polymorphisms are associated with the development of hepatocellular carcinoma in patients with chronic hepatitis B virus infection. Carcinogenesis. 2008;29:1192–6.CrossRefPubMed Yoon YJ, Chang HY, Ahn SH, et al. MDM2 and p53 polymorphisms are associated with the development of hepatocellular carcinoma in patients with chronic hepatitis B virus infection. Carcinogenesis. 2008;29:1192–6.CrossRefPubMed
19.
go back to reference Brown CJ, Cheok CF, Verma CS, et al. Reactivation of p53: from peptides to small molecules. Trends Pharmacol Sci. 2011;32:53–62.CrossRefPubMed Brown CJ, Cheok CF, Verma CS, et al. Reactivation of p53: from peptides to small molecules. Trends Pharmacol Sci. 2011;32:53–62.CrossRefPubMed
20.
22.
go back to reference Krecic AM, Swanson MS. hnRNP complexes: composition, structure, and function. Curr Opin Cell Biol. 1999;11(3):363–71.CrossRefPubMed Krecic AM, Swanson MS. hnRNP complexes: composition, structure, and function. Curr Opin Cell Biol. 1999;11(3):363–71.CrossRefPubMed
23.
go back to reference Kim MY, Hur J, Jeong S. Emerging roles of RNA and RNA-binding protein network in cancer cells. BMB Rep. 2009;42(3):125–30.CrossRefPubMed Kim MY, Hur J, Jeong S. Emerging roles of RNA and RNA-binding protein network in cancer cells. BMB Rep. 2009;42(3):125–30.CrossRefPubMed
24.
go back to reference Dreyfuss G, Matunis MJ, Pinol-Roma S, et al. hnRNP proteins and the biogenesis of mRNA. Annu Rev Biochem. 1993;62:289–321.CrossRefPubMed Dreyfuss G, Matunis MJ, Pinol-Roma S, et al. hnRNP proteins and the biogenesis of mRNA. Annu Rev Biochem. 1993;62:289–321.CrossRefPubMed
25.
26.
go back to reference Yisraeli JK. VICKZ proteins: a multi-talented family of regulatory RNA-binding proteins. Biol Cell. 2005;97(1):87–96.CrossRefPubMed Yisraeli JK. VICKZ proteins: a multi-talented family of regulatory RNA-binding proteins. Biol Cell. 2005;97(1):87–96.CrossRefPubMed
27.
28.
go back to reference Sonenberg N, Hinnebusch AG. New modes of translational control in development, behavior, and disease. Mol Cell. 2007;28(5):721–9.CrossRefPubMed Sonenberg N, Hinnebusch AG. New modes of translational control in development, behavior, and disease. Mol Cell. 2007;28(5):721–9.CrossRefPubMed
29.
go back to reference Zhang J, Jun Cho S, Chen X. RNPC1, an RNA-binding protein and a target of the p53 family, regulates p63 expression through mRNA stability. Proc Natl Acad Sci U S A. 2010;107(21):9614–9.CrossRefPubMedPubMedCentral Zhang J, Jun Cho S, Chen X. RNPC1, an RNA-binding protein and a target of the p53 family, regulates p63 expression through mRNA stability. Proc Natl Acad Sci U S A. 2010;107(21):9614–9.CrossRefPubMedPubMedCentral
30.
go back to reference Cho SJ, Zhang J, Chen X. RNPC1 modulates the RNA-binding activity of, and cooperates with, HuR to regulate p21 mRNA stability. Nucleic Acids Res. 2010;38(7):2256–67.CrossRefPubMedPubMedCentral Cho SJ, Zhang J, Chen X. RNPC1 modulates the RNA-binding activity of, and cooperates with, HuR to regulate p21 mRNA stability. Nucleic Acids Res. 2010;38(7):2256–67.CrossRefPubMedPubMedCentral
31.
go back to reference Yan W, Zhang J, Zhang Y, et al. p73 expression is regulated by RNPC1, a target of the p53 family, via mRNA stability. Mol Cell Biol. 2012;32(13):2336–48.CrossRefPubMedPubMedCentral Yan W, Zhang J, Zhang Y, et al. p73 expression is regulated by RNPC1, a target of the p53 family, via mRNA stability. Mol Cell Biol. 2012;32(13):2336–48.CrossRefPubMedPubMedCentral
32.
go back to reference Xu E, Zhang J, Chen X. MDM2 expression is repressed by the RNA-binding protein RNPC1 via mRNA stability. Oncogene. 2013;32(17):2169–78.CrossRefPubMed Xu E, Zhang J, Chen X. MDM2 expression is repressed by the RNA-binding protein RNPC1 via mRNA stability. Oncogene. 2013;32(17):2169–78.CrossRefPubMed
33.
go back to reference Zhang J, Cho SJ, Shu L, et al. Translational repression of p53 by RNPC1, a p53 targetoverexpressed in lymphomas. Genes Dev. 2001;25:1528–43.CrossRef Zhang J, Cho SJ, Shu L, et al. Translational repression of p53 by RNPC1, a p53 targetoverexpressed in lymphomas. Genes Dev. 2001;25:1528–43.CrossRef
34.
35.
go back to reference Zhang J, Xu E, Ren C, Cardiff DM, Imaie, Wisner E, Chen X, et al. Mice deficient in Rbm38, a target of the p53 family, are susceptible to accelerated aging and spontaneous tumors. PNAS. 2014;111(52):18637–42.CrossRefPubMed Zhang J, Xu E, Ren C, Cardiff DM, Imaie, Wisner E, Chen X, et al. Mice deficient in Rbm38, a target of the p53 family, are susceptible to accelerated aging and spontaneous tumors. PNAS. 2014;111(52):18637–42.CrossRefPubMed
36.
go back to reference Ding C, Cheng S, Yang Z, et al. Long non-coding RNA HOTAIR promotes cell migration and invasion via down-regulation of RNA binding motif protein 38 in hepatocellular carcinoma cells. Int J Mol Sci. 2014;15:4060–76. Ding C, Cheng S, Yang Z, et al. Long non-coding RNA HOTAIR promotes cell migration and invasion via down-regulation of RNA binding motif protein 38 in hepatocellular carcinoma cells. Int J Mol Sci. 2014;15:4060–76.
Metadata
Title
RBM38 plays a tumor-suppressor role via stabilizing the p53-mdm2 loop function in hepatocellular carcinoma
Authors
Jiazhou Ye
Rong Liang
Tao Bai
Yan Lin
Rongyun Mai
Meng Wei
Xinqin Ye
Lequn Li
Feixiang Wu
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2018
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0852-x

Other articles of this Issue 1/2018

Journal of Experimental & Clinical Cancer Research 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