RBMS2 Chemosensitizes Breast Cancer Cells to Doxorubicin by Regulating BMF Expression

Feng Xu, Tian Xia, Qi-Tong Xu, Xu Zhang, Yu-Zhou Huang, Xi Sun, Liang Shi, Xu-Jie Zhou, Ji-Fu Wei, Qiang Ding, Feng Xu, Tian Xia, Qi-Tong Xu, Xu Zhang, Yu-Zhou Huang, Xi Sun, Liang Shi, Xu-Jie Zhou, Ji-Fu Wei, Qiang Ding

Abstract

Chemoresistance is closely related to the therapeutic effect and prognosis in breast cancer patients. Increasing evidences demonstrated that RNA binding proteins (RBPs) have notable roles in regulating cancer cell proliferation, metastasis and chemotherapeutic sensitivity. RNA binding motif single stranded interacting protein 2 (RBMS2), an RBP, has been considered to be a tumor suppressor in several cancers. However, its role of doxorubicin sensitivity in breast cancer patients has not yet been fully revealed. Here, we performed doxorubicin cytotoxicity assay, flow cytometry and mouse xenograft model to examine the influence of RBMS2 on doxorubicin sensitization in vitro and in vivo. RIP assay and dual-luciferase reporter assay were performed to explore the relationship between RBMS2 and BMF. Our data demonstrated that upregulation of RBMS2 in breast cancer cells could enhance sensitivity to doxorubicin and promote apoptosis in the presence of doxorubicin, while inhibition of RBMS2 showed an opposite trend. Moreover, this chemosensitizing effect of RBMS2 could be reversed by the inhibition of Bcl-2 modifying factor (BMF). RBMS2 positively regulated BMF expression and increased BMF-induced expression of (cleaved) caspase 3, (cleaved) caspase 9 and poly (ADP-Ribose) polymerase (PARP). These results uncovered a novel mechanism for RBMS2 in the sensibilization of doxorubicin, suggesting that RBMS2 may act as a potential therapeutic target for drug-resistant breast cancer.

Keywords: BMF; RBMS2; apoptosis; breast cancer; chemosensitization; doxorubicin.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interest exists.

© The author(s).

Figures

Figure 1
Figure 1
RBMS2 could sensitize breast cancer cells to DOX in vitro. RBMS2 overexpression and knockdown cell lines were treated with different dosages of DOX for 24 h, respectively. CCK-8 assay was used to examine the cell viability (A and D for MCF-7, B and E for SUM 1315) and IC50 value (C and F) of DOX. Transfected MCF-7 (G, H) and SUM 1315 (I, J) cells were treated with DOX (0 or 1 µg/ml) for 2 weeks. Representative images (left panel) and quantification (right panel) of colonies in the colony formation assay. Data were shown as mean ± SD. *p <0.05, **p <0.01, ***p<0.001.
Figure 2
Figure 2
Overexpression of RBMS2 could enhance the therapeutic effect of DOX in vivo. Tumor volumes were measured in different treatment groups (A. B). The excised tumor lumps were weighed (C) and photographed (D). Data were shown as mean ± SD. *p <0.05, **p <0.01, ***p<0.001.
Figure 3
Figure 3
RBMS2 could induce apoptosis and apoptosis related proteins in breast cancer cell lines. Flow cytometry analysis of apoptosis in RBMS2 overexpression and knockdown MCF-7 and SUM 1315 cells with or without DOX treatment (A, B). The statistical calculation of apoptotic rates was shown (C). The levels of (cleaved) caspase 3, (cleaved) caspase 9 and PARP in MCF-7 and SUM 1315 cells transfected with RBMS2 overexpression and knockdown were detected by Western blot (D). The top band of RBMS2 is the exogenously band in western blot. Data were shown as mean ± SD. *p <0.05, **p <0.01, ***p<0.001.
Figure 4
Figure 4
RBMS2 regulated the expression of pro-apoptotic protein BMF. Volcano plot represented the distribution of mapped transcripts (A). The size and color of the dots meant the number of enriched genes and the adjusted p values, respectively. BMF was found to be positively correlated with RBMS2 in breast cancer both from TIMER (Tumor Immune Estimation Resource, https://cistrome.shinyapps.io/timer) database (B), the patients' samples from our hospital (C). Correlation heatmap were used to analyze the correlation between RBMS2 and BMF (D). R1, R2, R3 and V5-1, V5-2 and V5-3 represented RBMS2-overexpressed group and the control group, respectively. The red arrow indicated that BMF was positively correlated with RBMS2. Expression of BMF in breast cancer tissues and normal tissues (E, F). Overexpression of RBMS2 significantly increased the expression of BMF at both mRNA and protein (G, I) levels. BMF was significantly down regulated after RBMS2 knock down at both mRNA and protein levels (H, J). The top band of RBMS2 is the exogenously band in western blot. Data were shown as mean ± SD. *p <0.05, **p <0.01, ***p<0.001.
Figure 5
Figure 5
RBMS2 could increase BMF mRNA stability. In MCF-7 and SUM 1315 cell lines, RBMS2 overexpression prolonged the half-life of BMF mRNA after treated with Act D at a concentration of 5 µg/ml (A, B), while knockdown of RBMS2 shortened the half-life of BMF mRNA (C, D). MCF-7 and SUM 1315 cells lysates were immunoprecipitated with RBMS2 or IgG antibody and analyzed using PCR (E, G) and RT-qPCR (F, H) to detect transcript levels of BMF. Schematic diagram containing BMF 3'-UTR (upper) and AREs mutant region (lower) (I). The reporter containing BMF 3'-UTR was increased after overexpression of RBMS2 in MCF-7 (J) and SUM 1315 (K) cell lines. Firefly luciferase activity was detected and normalized to Renilla luciferase activity. Data were shown as mean ± SD. *p <0.05, **p <0.01, ***p<0.001.
Figure 6
Figure 6
BMF could reverse the sensitization to DOX induced by RBMS2 both in vivo and in vitro. Small interfering RNA of BMF was transfected into RBMS2 overexpression MCF- 7 and SUM 1315 cells. Transfection efficiency was confirmed via RT-qPCR and western blot (A, B). The top band of RBMS2 is the exogenously band in western blot. The sensitivity to DOX of MCF-7 and SUM 1315 cells mentioned above was examined using CCK-8 and colony formation. CCK-8 assay was used to examine cell viability (C for MCF-7, D for SUM 1315) and IC50 value (E) of DOX. Representative photographs (F, I), quantification (G, J) and cell survival after DOX treatment (H, K) were shown by colony formation assay. Tumor volumes were measured in different treatment groups (L). The excised tumor lumps were weighed (M) and photographed (N). Data were shown as mean ± SD. *p <0.05, **p <0.01, ***p<0.001.
Figure 7
Figure 7
BMF could rescue the apoptosis and apoptosis related proteins induced by RBMS2 in breast cancer cell lines. Apoptosis and apoptosis related proteins were detected by flow cytometry analysis (A, B, C) and western blot (D, E). Data were shown as mean ± SD. *p <0.05, **p <0.01, ***p<0.001.

References

    1. Fan L, Strasser-Weippl K, Li J, St Louis J, Finkelstein D, Yu K. et al. Breast cancer in China. The Lancet Oncology. 2014;15:e279–89.
    1. Siegel R, Miller K, Jemal A. Cancer statistics, 2020. CA: a cancer journal for clinicians. 2020;70:7–30.
    1. Ma X, Wang M, Yin T, Zhao Y, Wei X. Myeloid-Derived Suppressor Cells Promote Metastasis in Breast Cancer After the Stress of Operative Removal of the Primary Cancer. Frontiers in oncology. 2019;9:855.
    1. Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival. an overview of the randomised trials. Lancet (London, England) 2005;365:1687–717.
    1. Wang C, Jin H, Wang N, Fan S, Wang Y, Zhang Y. et al. Gas6/Axl Axis Contributes to Chemoresistance and Metastasis in Breast Cancer through Akt/GSK-3β/β-catenin Signaling. Theranostics. 2016;6:1205–19.
    1. Liu X, Zheng J, Sun W, Zhao X, Li Y, Gong N. et al. Ferrimagnetic Vortex Nanoring-Mediated Mild Magnetic Hyperthermia Imparts Potent Immunological Effect for Treating Cancer Metastasis. ACS Nano. 2019;13:8811–25.
    1. Ji X, Lu Y, Tian H, Meng X, Wei M, Cho W. Chemoresistance mechanisms of breast cancer and their countermeasures. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie. 2019;114:108800.
    1. Venkatadri R, Muni T, Iyer A, Yakisich J, Azad N. Role of apoptosis-related miRNAs in resveratrol-induced breast cancer cell death. Cell death & disease. 2016;7:e2104.
    1. Pilco-Ferreto N, Calaf G. Influence of doxorubicin on apoptosis and oxidative stress in breast cancer cell lines. International journal of oncology. 2016;49:753–62.
    1. Nitiss J. Targeting DNA topoisomerase II in cancer chemotherapy. Nature reviews Cancer. 2009;9:338–50.
    1. Gradishar W, Anderson B, Abraham J, Aft R, Agnese D, Allison K. et al. Breast Cancer, Version 3.2020, NCCN Clinical Practice Guidelines in Oncology. Journal of the National Comprehensive Cancer Network: JNCCN. 2020;18:452–78.
    1. Zeng H, Wang L, Wang J, Chen T, Li H, Zhang K. et al. microRNA-129-5p suppresses Adriamycin resistance in breast cancer by targeting SOX2. Archives of biochemistry and biophysics. 2018;651:52–60.
    1. Liu J, Debnath J. The Evolving, Multifaceted Roles of Autophagy in Cancer. Advances in cancer research. 2016;130:1–53.
    1. Balaji S, Udupa N, Chamallamudi M, Gupta V, Rangarajan A. Role of the Drug Transporter ABCC3 in Breast Cancer Chemoresistance. PloS one. 2016;11:e0155013.
    1. Thomas S, Quinn B, Das S, Dash R, Emdad L, Dasgupta S. et al. Targeting the Bcl-2 family for cancer therapy. Expert opinion on therapeutic targets. 2013;17:61–75.
    1. Mortezaee K, Salehi E, Mirtavoos-Mahyari H, Motevaseli E, Najafi M, Farhood B. et al. Mechanisms of apoptosis modulation by curcumin: Implications for cancer therapy. Journal of cellular physiology. 2019;234:12537–50.
    1. Kurppa K, Liu Y, To C, Zhang T, Fan M, Vajdi A. et al. Treatment-Induced Tumor Dormancy through YAP-Mediated Transcriptional Reprogramming of the Apoptotic Pathway. Cancer cell. 2020;37:104–22.e12.
    1. 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. Journal of experimental & clinical cancer research: CR. 2017;36:49.
    1. Pereira B, Billaud M, Almeida R. RNA-Binding Proteins in Cancer: Old Players and New Actors. Trends in cancer. 2017;3:506–28.
    1. Panzeri V, Manni I, Capone A, Naro C, Sacconi A, Di Agostino S. et al. The RNA-binding protein MEX3A is a prognostic factor and regulator of resistance to gemcitabine in pancreatic ductal adenocarcinoma. Molecular oncology. 2021;15:579–95.
    1. Wang H, Huang R, Guo W, Qin X, Yang Z, Yuan Z. et al. RNA-binding protein CELF1 enhances cell migration, invasion, and chemoresistance by targeting ETS2 in colorectal cancer. Clinical science (London, England: 1979) 2020;134:1973–90.
    1. Kim S, Ju J, Kang M, Eun J, Kim Y, Raninga P. et al. RNA-binding protein NONO contributes to cancer cell growth and confers drug resistance as a theranostic target in TNBC. Theranostics. 2020;10:7974–92.
    1. Kanaoka Y, Nojima H. SCR: novel human suppressors of cdc2/cdc13 mutants of Schizosaccharomyces pombe harbour motifs for RNA binding proteins. Nucleic acids research. 1994;22:2687–93.
    1. Sun X, Hu Y, Wu J, Shi L, Zhu L, Xi P. et al. RBMS2 inhibits the proliferation by stabilizing P21 mRNA in breast cancer. Journal of experimental & clinical cancer research: CR. 2018;37:298.
    1. Burke P. Mitochondria, Bioenergetics and Apoptosis in Cancer. Trends in cancer. 2017;3:857–70.
    1. Carneiro B, El-Deiry W. Targeting apoptosis in cancer therapy. Nature reviews Clinical oncology. 2020;17:395–417.
    1. Zhu L, Xi P, Li X, Sun X, Zhou W, Xia T. et al. The RNA binding protein RBMS3 inhibits the metastasis of breast cancer by regulating Twist1 expression. Journal of experimental & clinical cancer research: CR. 2019;38:105.
    1. Xi P, Zhang X, Zhu L, Dai X, Cheng L, Hu Y. et al. Oncogenic action of the exosome cofactor RBM7 by stabilization of CDK1 mRNA in breast cancer. NPJ breast cancer. 2020;6:58.
    1. 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. British journal of cancer. 2017;117:675–84.
    1. Tianhu Z, Shiguang Z, Xinghan L. Bmf is upregulated by PS-341-mediated cell death of glioma cells through JNK phosphorylation. Molecular biology reports. 2010;37:1211–9.
    1. Zhang Y, Adachi M, Kawamura R, Zou H, Imai K, Hareyama M. et al. Bmf contributes to histone deacetylase inhibitor-mediated enhancing effects on apoptosis after ionizing radiation. Apoptosis: an international journal on programmed cell death. 2006;11:1349–57.
    1. Hinds M, Smits C, Fredericks-Short R, Risk J, Bailey M, Huang D. et al. Bim, Bad and Bmf: intrinsically unstructured BH3-only proteins that undergo a localized conformational change upon binding to prosurvival Bcl-2 targets. Cell death and differentiation. 2007;14:128–36.
    1. Kutuk O, Letai A. Displacement of Bim by Bmf and Puma rather than increase in Bim level mediates paclitaxel-induced apoptosis in breast cancer cells. Cell death and differentiation. 2010;17:1624–35.
    1. Tan B, Tiong K, Choo H, Chung F, Hii L, Tan S. et al. Mutant p53-R273H mediates cancer cell survival and anoikis resistance through AKT-dependent suppression of BCL2-modifying factor (BMF) Cell death & disease. 2015;6:e1826.
    1. Xue J, Xia T, Liang X, Zhou W, Cheng L, Shi L. et al. RNA-binding protein RNPC1: acting as a tumor suppressor in breast cancer. BMC cancer. 2014;14:322.
    1. 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. Journal of experimental & clinical cancer research: CR. 2017;36:149.

Source: PubMed

3
Abonnieren