Skip to main content
Top
Published in: Breast Cancer Research 1/2019

Open Access 01-12-2019 | Metastasis | Research article

Context-dependent roles of MDMX (MDM4) and MDM2 in breast cancer proliferation and circulating tumor cells

Authors: Chong Gao, Gu Xiao, Alessandra Piersigilli, Jiangtao Gou, Olorunseun Ogunwobi, Jill Bargonetti

Published in: Breast Cancer Research | Issue 1/2019

Login to get access

Abstract

Introduction

Many human breast cancers overexpress the E3 ubiquitin ligase MDM2 and its homolog MDMX. Expression of MDM2 and MDMX occurs in estrogen receptor α-positive (ERα+) breast cancer and triple-negative breast cancer (TNBC). There are p53-independent influences of MDM2 and MDMX, and 80% of TNBC express mutant p53 (mtp53). MDM2 drives TNBC circulating tumor cells (CTCs) in mice, but the context-dependent influences of MDM2 and MDMX on different subtypes of breast cancers expressing mtp53 have not been determined.

Methods

To assess the context-dependent roles, we carried out MDM2 and MDMX knockdown in orthotopic tumors of TNBC MDA-MB-231 cells expressing mtp53 R280K and MDM2 knockdown in ERα+ T47D cells expressing mtp53 L194F. The corresponding cell proliferation was scored in vitro by growth curves and in vivo by orthotopic tumor volumes. Cell migration was assessed in vitro by wound-healing assays and cell intravasation in vivo by sorting GFP-positive CTCs by flow cytometry. The metastasis gene targets were determined by an RT-PCR array card screen and verified by qRT-PCR and Western blot analysis.

Results

Knocking down MDMX or MDM2 in MDA-MB-231 cells reduced cell migration and CTC detection, but only MDMX knockdown reduced tumor volumes at early time points. This is the first report of MDMX overexpression in TNBC enhancing the CTC phenotype with correlated upregulation of CXCR4. Experiments were carried out to compare MDM2-knockdown outcomes in nonmetastatic ERα+ T47D cells. The knockdown of MDM2 in ERα+ T47D orthotopic tumors decreased primary tumor volumes, supporting our previous finding that estrogen-activated MDM2 increases cell proliferation.

Conclusions

This is the first report showing that the expression of MDM2 in ERα+ breast cancer and TNBC can result in different tumor-promoting outcomes. Both MDMX and MDM2 overexpression in TNBC MDA-MB-231 cells enhanced the CTC phenotype. These data indicate that both MDM2 and MDMX can promote TNBC metastasis and that it is important to consider the context-dependent roles of MDM2 family members in different subtypes of breast cancer.
Appendix
Available only for authorised users
Literature
1.
go back to reference Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70.CrossRef Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70.CrossRef
2.
3.
go back to reference Turbin DA, et al. MDM2 protein expression is a negative prognostic marker in breast carcinoma. Mod Pathol. 2006;19(1):69–74.PubMedCrossRef Turbin DA, et al. MDM2 protein expression is a negative prognostic marker in breast carcinoma. Mod Pathol. 2006;19(1):69–74.PubMedCrossRef
6.
go back to reference Brekman A, et al. A p53-independent role of Mdm2 in estrogen-mediated activation of breast cancer cell proliferation. Breast Cancer Res. 2011;13(1):R3.PubMedPubMedCentralCrossRef Brekman A, et al. A p53-independent role of Mdm2 in estrogen-mediated activation of breast cancer cell proliferation. Breast Cancer Res. 2011;13(1):R3.PubMedPubMedCentralCrossRef
7.
8.
go back to reference Migliorini D, et al. Hdmx recruitment into the nucleus by Hdm2 is essential for its ability to regulate p53 stability and transactivation. J Biol Chem. 2002;277(9):7318–23.PubMedCrossRef Migliorini D, et al. Hdmx recruitment into the nucleus by Hdm2 is essential for its ability to regulate p53 stability and transactivation. J Biol Chem. 2002;277(9):7318–23.PubMedCrossRef
9.
go back to reference Tanimura S, et al. MDM2 interacts with MDMX through their RING finger domains. FEBS Lett. 1999;447(1):5–9.PubMedCrossRef Tanimura S, et al. MDM2 interacts with MDMX through their RING finger domains. FEBS Lett. 1999;447(1):5–9.PubMedCrossRef
10.
go back to reference Pan Y, Chen J. MDM2 promotes ubiquitination and degradation of MDMX. Mol Cell Biol. 2003;23(15):5113–5121. Pan Y, Chen J. MDM2 promotes ubiquitination and degradation of MDMX. Mol Cell Biol. 2003;23(15):5113–5121.
11.
12.
go back to reference Haupt S, et al. The role of MDM2 and MDM4 in breast cancer development and prevention. J Mol Cell Biol. 2017;9(1):53–61.PubMedPubMedCentral Haupt S, et al. The role of MDM2 and MDM4 in breast cancer development and prevention. J Mol Cell Biol. 2017;9(1):53–61.PubMedPubMedCentral
14.
go back to reference Lu X, et al. Mouse double minute 2 (MDM2) upregulates Snail expression and induces epithelial-to-mesenchymal transition in breast cancer cells in vitro and in vivo. Oncotarget. 2016;7(24):37177–91.PubMedPubMedCentral Lu X, et al. Mouse double minute 2 (MDM2) upregulates Snail expression and induces epithelial-to-mesenchymal transition in breast cancer cells in vitro and in vivo. Oncotarget. 2016;7(24):37177–91.PubMedPubMedCentral
15.
go back to reference Chen X, et al. MDM2 promotes invasion and metastasis in invasive ductal breast carcinoma by inducing matrix metalloproteinase-9. PLoS One. 2013;8(11):e78794.PubMedPubMedCentralCrossRef Chen X, et al. MDM2 promotes invasion and metastasis in invasive ductal breast carcinoma by inducing matrix metalloproteinase-9. PLoS One. 2013;8(11):e78794.PubMedPubMedCentralCrossRef
16.
go back to reference Slabakova E, et al. Opposite regulation of MDM2 and MDMX expression in acquisition of mesenchymal phenotype in benign and cancer cells. Oncotarget. 2015;6(34):36156–71.PubMedPubMedCentralCrossRef Slabakova E, et al. Opposite regulation of MDM2 and MDMX expression in acquisition of mesenchymal phenotype in benign and cancer cells. Oncotarget. 2015;6(34):36156–71.PubMedPubMedCentralCrossRef
18.
go back to reference Miranda PJ, et al. MDM4 is a rational target for treating breast cancers with mutant p53. J Pathol. 2017;241(5):661–70.PubMedCrossRef Miranda PJ, et al. MDM4 is a rational target for treating breast cancers with mutant p53. J Pathol. 2017;241(5):661–70.PubMedCrossRef
22.
go back to reference Kang Y, et al. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell. 2003;3(6):537–49.PubMedCrossRef Kang Y, et al. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell. 2003;3(6):537–49.PubMedCrossRef
24.
go back to reference Pfister NT, et al. Mutant p53 cooperates with the SWI/SNF chromatin remodeling complex to regulate VEGFR2 in breast cancer cells. Genes Dev. 2015;29(12):1298–315.PubMedPubMedCentralCrossRef Pfister NT, et al. Mutant p53 cooperates with the SWI/SNF chromatin remodeling complex to regulate VEGFR2 in breast cancer cells. Genes Dev. 2015;29(12):1298–315.PubMedPubMedCentralCrossRef
25.
go back to reference Shapiro SS, Wilk MB. An analysis of variance test for normality (complete samples). Biometrika. 1965;52:591–611.CrossRef Shapiro SS, Wilk MB. An analysis of variance test for normality (complete samples). Biometrika. 1965;52:591–611.CrossRef
26.
go back to reference Gou JT, et al. A class of improved hybrid Hochberg-Hommel type step-up multiple test procedures. Biometrika. 2014;101(4):899–911.CrossRef Gou JT, et al. A class of improved hybrid Hochberg-Hommel type step-up multiple test procedures. Biometrika. 2014;101(4):899–911.CrossRef
27.
go back to reference Hochberg Y. A sharper Bonferroni procedure for multiple tests of significance. Biometrika. 1988;75(4):800–2.CrossRef Hochberg Y. A sharper Bonferroni procedure for multiple tests of significance. Biometrika. 1988;75(4):800–2.CrossRef
28.
go back to reference Tamhane AC, Gou J. Advances in p-value based multiple test procedures. J Biopharm Stat. 2018;28(1):10–27.PubMedCrossRef Tamhane AC, Gou J. Advances in p-value based multiple test procedures. J Biopharm Stat. 2018;28(1):10–27.PubMedCrossRef
30.
go back to reference Kenny PA, et al. The morphologies of breast cancer cell lines in three-dimensional assays correlate with their profiles of gene expression. Mol Oncol. 2007;1(1):84–96.PubMedPubMedCentralCrossRef Kenny PA, et al. The morphologies of breast cancer cell lines in three-dimensional assays correlate with their profiles of gene expression. Mol Oncol. 2007;1(1):84–96.PubMedPubMedCentralCrossRef
32.
go back to reference Li S, Huang S, Peng SB. Overexpression of G protein-coupled receptors in cancer cells: involvement in tumor progression. Int J Oncol. 2005;27(5):1329–39.PubMed Li S, Huang S, Peng SB. Overexpression of G protein-coupled receptors in cancer cells: involvement in tumor progression. Int J Oncol. 2005;27(5):1329–39.PubMed
34.
go back to reference Arun B, Goss P. The role of COX-2 inhibition in breast cancer treatment and prevention. Semin Oncol. 2004;31(2 Suppl 7):22–29. Arun B, Goss P. The role of COX-2 inhibition in breast cancer treatment and prevention. Semin Oncol. 2004;31(2 Suppl 7):22–29.
35.
go back to reference Chen JY, et al. Cancer/stroma interplay via cyclooxygenase-2 and indoleamine 2,3-dioxygenase promotes breast cancer progression. Breast Cancer Res. 2014;16(4):410.PubMedPubMedCentralCrossRef Chen JY, et al. Cancer/stroma interplay via cyclooxygenase-2 and indoleamine 2,3-dioxygenase promotes breast cancer progression. Breast Cancer Res. 2014;16(4):410.PubMedPubMedCentralCrossRef
36.
go back to reference Fakharzadeh SS, Trusko SP, George DL. Tumorigenic potential associated with enhanced expression of a gene that is amplified in a mouse tumor cell line. EMBO J. 1991;10(6):1565–9.PubMedPubMedCentralCrossRef Fakharzadeh SS, Trusko SP, George DL. Tumorigenic potential associated with enhanced expression of a gene that is amplified in a mouse tumor cell line. EMBO J. 1991;10(6):1565–9.PubMedPubMedCentralCrossRef
37.
go back to reference Danovi D, et al. Amplification of Mdmx (or Mdm4) directly contributes to tumor formation by inhibiting p53 tumor suppressor activity. Mol Cell Biol. 2004;24(13):5835–43.PubMedPubMedCentralCrossRef Danovi D, et al. Amplification of Mdmx (or Mdm4) directly contributes to tumor formation by inhibiting p53 tumor suppressor activity. Mol Cell Biol. 2004;24(13):5835–43.PubMedPubMedCentralCrossRef
39.
go back to reference Jones SN, et al. Overexpression of Mdm2 in mice reveals a p53-independent role for Mdm2 in tumorigenesis. Proc Natl Acad Sci U S A. 1998;95(26):15608–12.PubMedPubMedCentralCrossRef Jones SN, et al. Overexpression of Mdm2 in mice reveals a p53-independent role for Mdm2 in tumorigenesis. Proc Natl Acad Sci U S A. 1998;95(26):15608–12.PubMedPubMedCentralCrossRef
42.
go back to reference De Clercq S, et al. Widespread overexpression of epitope-tagged Mdm4 does not accelerate tumor formation in vivo. Mol Cell Biol. 2010;30(22):5394–405.PubMedPubMedCentralCrossRef De Clercq S, et al. Widespread overexpression of epitope-tagged Mdm4 does not accelerate tumor formation in vivo. Mol Cell Biol. 2010;30(22):5394–405.PubMedPubMedCentralCrossRef
43.
go back to reference Hench IB, Hench J, Tolnay M. Liquid biopsy in clinical management of breast, lung, and colorectal cancer. Front Med (Lausanne). 2018;5:9.CrossRef Hench IB, Hench J, Tolnay M. Liquid biopsy in clinical management of breast, lung, and colorectal cancer. Front Med (Lausanne). 2018;5:9.CrossRef
44.
go back to reference Zhao H, et al. CXCR4 over-expression and survival in cancer: a system review and meta-analysis. Oncotarget. 2015;6(7):5022–40.PubMedCrossRef Zhao H, et al. CXCR4 over-expression and survival in cancer: a system review and meta-analysis. Oncotarget. 2015;6(7):5022–40.PubMedCrossRef
45.
go back to reference Pernas S, et al. Balixafortide plus eribulin in HER2-negative metastatic breast cancer: a phase 1, single-arm, dose-escalation trial. Lancet Oncol. 2018;19(6):812–24.PubMedCrossRef Pernas S, et al. Balixafortide plus eribulin in HER2-negative metastatic breast cancer: a phase 1, single-arm, dose-escalation trial. Lancet Oncol. 2018;19(6):812–24.PubMedCrossRef
46.
go back to reference Balkwill F. The significance of cancer cell expression of the chemokine receptor CXCR4. Semin Cancer Biol. 2004;14(3):171–9.PubMedCrossRef Balkwill F. The significance of cancer cell expression of the chemokine receptor CXCR4. Semin Cancer Biol. 2004;14(3):171–9.PubMedCrossRef
47.
go back to reference Muller A, et al. Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001;410(6824):50–6.PubMedCrossRef Muller A, et al. Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001;410(6824):50–6.PubMedCrossRef
48.
go back to reference Saha A, et al. Proinflammatory CXCL12-CXCR4/CXCR7 signaling axis drives Myc-induced prostate cancer in obese mice. Cancer Res. 2017;77(18):5158–68.PubMedPubMedCentral Saha A, et al. Proinflammatory CXCL12-CXCR4/CXCR7 signaling axis drives Myc-induced prostate cancer in obese mice. Cancer Res. 2017;77(18):5158–68.PubMedPubMedCentral
49.
go back to reference Ishikawa T, et al. Hypoxia enhances CXCR4 expression by activating HIF-1 in oral squamous cell carcinoma. Oncol Rep. 2009;21(3):707–12.PubMed Ishikawa T, et al. Hypoxia enhances CXCR4 expression by activating HIF-1 in oral squamous cell carcinoma. Oncol Rep. 2009;21(3):707–12.PubMed
50.
go back to reference Zagzag D, et al. Hypoxia-inducible factor 1 and VEGF upregulate CXCR4 in glioblastoma: implications for angiogenesis and glioma cell invasion. Lab Investig. 2006;86(12):1221–32.PubMedCrossRef Zagzag D, et al. Hypoxia-inducible factor 1 and VEGF upregulate CXCR4 in glioblastoma: implications for angiogenesis and glioma cell invasion. Lab Investig. 2006;86(12):1221–32.PubMedCrossRef
51.
go back to reference Helbig G, et al. NF-κB promotes breast cancer cell migration and metastasis by inducing the expression of the chemokine receptor CXCR4. J Biol Chem. 2003;278(24):21631–8.PubMedCrossRef Helbig G, et al. NF-κB promotes breast cancer cell migration and metastasis by inducing the expression of the chemokine receptor CXCR4. J Biol Chem. 2003;278(24):21631–8.PubMedCrossRef
52.
go back to reference Gu L, Findley HW, Zhou M. MDM2 induces NF-κB/p65 expression transcriptionally through Sp1-binding sites: a novel, p53-independent role of MDM2 in doxorubicin resistance in acute lymphoblastic leukemia. Blood. 2002;99(9):3367–75.PubMedCrossRef Gu L, Findley HW, Zhou M. MDM2 induces NF-κB/p65 expression transcriptionally through Sp1-binding sites: a novel, p53-independent role of MDM2 in doxorubicin resistance in acute lymphoblastic leukemia. Blood. 2002;99(9):3367–75.PubMedCrossRef
53.
go back to reference Vaughan C, et al. Human oncoprotein MDM2 up-regulates expression of NF-κB2 precursor p100 conferring a survival advantage to lung cells. Genes Cancer. 2011;2(10):943–55.PubMedPubMedCentralCrossRef Vaughan C, et al. Human oncoprotein MDM2 up-regulates expression of NF-κB2 precursor p100 conferring a survival advantage to lung cells. Genes Cancer. 2011;2(10):943–55.PubMedPubMedCentralCrossRef
54.
go back to reference Qiu WG, et al. Identification, validation, and targeting of the mutant p53-PARP-MCM chromatin axis in triple negative breast cancer. NPJ Breast Cancer. 2017;3:1. Qiu WG, et al. Identification, validation, and targeting of the mutant p53-PARP-MCM chromatin axis in triple negative breast cancer. NPJ Breast Cancer. 2017;3:1.
Metadata
Title
Context-dependent roles of MDMX (MDM4) and MDM2 in breast cancer proliferation and circulating tumor cells
Authors
Chong Gao
Gu Xiao
Alessandra Piersigilli
Jiangtao Gou
Olorunseun Ogunwobi
Jill Bargonetti
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Breast Cancer Research / Issue 1/2019
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-018-1094-8

Other articles of this Issue 1/2019

Breast 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