1
|
Feldheim J, Kessler AF, Feldheim JJ, Schmitt D, Oster C, Lazaridis L, Glas M, Ernestus RI, Monoranu CM, Löhr M, Hagemann C. BRMS1 in Gliomas-An Expression Analysis. Cancers (Basel) 2023; 15:cancers15112907. [PMID: 37296870 DOI: 10.3390/cancers15112907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
The metastatic suppressor BRMS1 interacts with critical steps of the metastatic cascade in many cancer entities. As gliomas rarely metastasize, BRMS1 has mainly been neglected in glioma research. However, its interaction partners, such as NFκB, VEGF, or MMPs, are old acquaintances in neurooncology. The steps regulated by BRMS1, such as invasion, migration, and apoptosis, are commonly dysregulated in gliomas. Therefore, BRMS1 shows potential as a regulator of glioma behavior. By bioinformatic analysis, in addition to our cohort of 118 specimens, we determined BRMS1 mRNA and protein expression as well as its correlation with the clinical course in astrocytomas IDH mutant, CNS WHO grade 2/3, and glioblastoma IDH wild-type, CNS WHO grade 4. Interestingly, we found BRMS1 protein expression to be significantly decreased in the aforementioned gliomas, while BRMS1 mRNA appeared to be overexpressed throughout. This dysregulation was independent of patients' characteristics or survival. The protein and mRNA expression differences cannot be finally explained at this stage. However, they suggest a post-transcriptional dysregulation that has been previously described in other cancer entities. Our analyses present the first data on BRMS1 expression in gliomas that can provide a starting point for further investigations.
Collapse
Affiliation(s)
- Jonas Feldheim
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, 45131 Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Hufelandstraße 55, 45131 Essen, Germany
| | - Almuth F Kessler
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
| | - Julia J Feldheim
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
- Department of Neurosurgery, University Hospital Essen, Hufelandstraße 55, 45131 Essen, Germany
| | - Dominik Schmitt
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
- Department of Nuclear Medicine, University of Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Christoph Oster
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, 45131 Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Hufelandstraße 55, 45131 Essen, Germany
| | - Lazaros Lazaridis
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, 45131 Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Hufelandstraße 55, 45131 Essen, Germany
| | - Martin Glas
- Division of Clinical Neurooncology, Department of Neurology, University Hospital Essen, University Duisburg-Essen, Hufelandstraße 55, 45131 Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Hufelandstraße 55, 45131 Essen, Germany
| | - Ralf-Ingo Ernestus
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
| | - Camelia M Monoranu
- Department of Neuropathology, Institute of Pathology, University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Mario Löhr
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
| | - Carsten Hagemann
- Section Experimental Neurosurgery, Department of Neurosurgery, University of Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
| |
Collapse
|
2
|
Babu D, Chintal R, Panigrahi M, Phanithi PB. Distinct expression and function of breast cancer metastasis suppressor 1 in mutant P53 glioblastoma. Cell Oncol (Dordr) 2022; 45:1451-1465. [PMID: 36284039 DOI: 10.1007/s13402-022-00729-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2022] [Indexed: 01/10/2023] Open
Abstract
PURPOSE Glioblastoma (GBM) is the most malignant subtype of astrocytic tumors with the worst prognosis in all its progressive forms. Breast cancer metastasis suppressor 1 (BRMS1) is a metastasis suppressor gene that controls malignancy in multiple tumors. As yet, however, its clinical and functional significance in mutant P53 GBM remains inconclusive. Here, we attempted to study the importance of BRMS1 in mutant P53 GBM. METHODS BRMS1 expression was evaluated in 74 human astrocytoma tissues by qRT-PCR, Western blotting and immunohistochemistry. BRMS1 expression in the astrocytoma tissues was correlated with clinicopathological parameters, the P53 mutation status and BRMS1 downstream targets, and compared with TCGA and NCI-60 datasets. siRNA-mediated knockdown of BRMS1 was performed in selected GBM cell lines to evaluate the functional role of BRMS1. RESULTS Our study revealed an enhanced expression of BRMS1 in GBM which was associated with a poor patient survival, and this observation was corroborated by the TCGA dataset. We also found a positive correlation between BRMS1 expression and a mutant P53 status in GBM which was associated with a poor prognosis. In vitro BRMS1 silencing reduced the growth of mutant P53 GBM cells and repressed their colonization and migration/invasion by modulating EGFR-AKT/NF-κB signaling. Transcriptional profiling revealed a positive and negative correlation of uPA and ING4 expression with BRMS1 expression, respectively. CONCLUSION Our data indicate upregulation of BRMS1 in high grade astrocytomas which correlates positively with mutant P53 and a poor patient survival. Silencing of BRMS1 in mutant P53 GBM cell lines resulted in a reduced cellular growth and migration/invasion by suppressing the EGFR-AKT/NF-kB signaling pathway. BRMS1 may serve as a predictive biomarker and therapeutic target in mutant P53 GBM.
Collapse
Affiliation(s)
- Deepak Babu
- Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Room No: F-23/F-71, Hyderabad, Telangana State, 500 046, India
| | - Ramulu Chintal
- Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Room No: F-23/F-71, Hyderabad, Telangana State, 500 046, India
| | - Manas Panigrahi
- Department of Neurosurgery, Krishna Institute of Medical Sciences, 500 003, Secunderabad, Telangana State, India
| | - Prakash Babu Phanithi
- Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Room No: F-23/F-71, Hyderabad, Telangana State, 500 046, India.
| |
Collapse
|
3
|
Farokhimanesh S, Forouzandeh Moghadam M, Ebrahimi M, Hashemi ZS. Metastasis Inhibition by Cell Type Specific Expression of BRMS1 Gene under The Regulation of miR200 Family Response Elements. CELL JOURNAL 2021; 23:225-237. [PMID: 34096224 PMCID: PMC8181311 DOI: 10.22074/cellj.2021.6988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 10/03/2019] [Indexed: 11/23/2022]
Abstract
Objective Specific expression of therapeutic genes in cancer therapy has been per used for many years. One of the
innovative strategies that have recently been introduced is employing miRNA response elements (MREs) of microRNAs
(whose expression are reduced or inhibited in cancerous cells) into the 3´UTR of the therapeutic genes for their specific
expression. Accordingly, MREs of anti-metastatic miRNA family have been used in 3´UTR of the metastasis suppressor
gene in the corresponding cells to evaluate the level of metastatic behavior. Material and Methods In this experimental study, 3´UTR of the ZEB1 gene with 592 bp length, encompassing multiple
MREs of miR-141, miR-429, miR-200b and miR-200c, was employed to replace BRMS1 3´UTR. The obtained vector
was then assessed in the context of MCF-10A, MDA-MB231 and MCF-7 cells. Results It was shown that the employed MREs are able to up-regulate BRMS expression in the metastatic MDA-
MB231 cells (almost 3.5-fold increase), while it was significantly reduced within tumorigenic/non-metastatic MCF-7
cells. Specific expression of BRMS1 in metastatic cells led to a significant reduction in their migratory and invasive
characteristics (about 65% and 55%, respectively). Two-tailed student’s t test was utilized for statistical analysis. Conclusion It was demonstrated that a chimeric vector containing BRMS1 which is regulated by miR-200 family
response element may represent a promising therapeutic tool. This is due to the capability of the chimeric vector for
cell type-specific expression of anti-metastatic genes with lowest side-effects. It consequently prohibits the invasive
characteristics of metastatic cells.
Collapse
Affiliation(s)
- Samila Farokhimanesh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Department of Biotechnology, Applied Biophotonics Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mahdi Forouzandeh Moghadam
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Sadat Hashemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Abstract
Despite high mortality rates, molecular understanding of metastasis remains limited. It can be regulated by both pro- and anti-metastasis genes. The metastasis suppressor, breast cancer metastasis suppressor 1 (BRMS1), has been positively correlated with patient outcomes, but molecular functions are still being characterized. BRMS1 has been implicated in focal adhesion kinase (FAK), epidermal growth factor receptor (EGFR), and NF-κB signaling pathways. We review evidence that BRMS1 regulates these vast signaling pathways through chromatin remodeling as a member of mSin3 histone deacetylase complexes.
Collapse
|
5
|
Du Y, Yang F, Lv D, Zhang Q, Yuan X. MiR-147 inhibits cyclic mechanical stretch-induced apoptosis in L6 myoblasts via ameliorating endoplasmic reticulum stress by targeting BRMS1. Cell Stress Chaperones 2019; 24:1151-1161. [PMID: 31628639 PMCID: PMC6882977 DOI: 10.1007/s12192-019-01037-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/16/2019] [Accepted: 09/23/2019] [Indexed: 01/11/2023] Open
Abstract
Functional orthopedic treatment is effective for the correction of malformation. Studies demonstrated myoblasts undergo proliferation and apoptosis on certain stretch conditions. MicroRNAs (miRNAs) function in RNA silencing and post-transcriptional regulation of gene expression, and participate in various biological processes, including proliferation and apoptosis. One hypothesis suggested that miRNA was involved into the procedure via suppressing its target genes then triggered endoplasmic reticulum stress-induced apoptosis. Therefore, miRNAs play important roles in the regulation of the proliferation and apoptosis of myoblasts. In our study, the miR-147 has been explored. A cyclic mechanical stretch model was established to observe the features of rat L6 myoblasts. The detection of mRNA and protein levels was performed by qRT-PCR and western blot. L6 cell proliferation/apoptosis was checked by CCK-8 assay, DNA fragmentation assay, and caspase-3 activity assay. MiRNA transfections were performed as per the manufacturer's suggestions: (1) cyclic mechanical stretch induced apoptosis of L6 myoblasts and inhibition of miR-147; (2) miR-147 attenuated cyclic mechanical stretch-induced apoptosis of L6 myoblasts; (3) miR-147 attenuated cyclic mechanical stretch-induced L6 myoblast endoplasmic reticulum stress; (4) BRMS1 was a direct target of miR-147 in L6 myoblasts; (5) miR-147/BRMS1 axis participated in the regulation of cyclic mechanical stress on L6 myoblasts. MiR-147 attenuates endoplasmic reticulum stress by targeting BRMS1 to inhibit cyclic mechanical stretch-induced apoptosis of L6 myoblasts.
Collapse
Affiliation(s)
- Yanxiao Du
- Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Stomatology, Qingdao Central Hospital, Qingdao, 266042, Shandong, China
| | - Feng Yang
- School of Stomatology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Department of Stomatology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Di Lv
- Department of Stomatology, Qingdao Central Hospital, Qingdao, 266042, Shandong, China
| | - Qiang Zhang
- Department of Orthodontics II, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Xiao Yuan
- Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- Department of Orthodontics II, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China.
| |
Collapse
|
6
|
Cao Y, Tan S, Tu Y, Zhang G, Liu Y, Li D, Xu S, Le Z, Xiong J, Zou W, Gong P, Li Z, Jie Z. MicroRNA-125a-5p inhibits invasion and metastasis of gastric cancer cells by targeting BRMS1 expression. Oncol Lett 2018; 15:5119-5130. [PMID: 29552146 DOI: 10.3892/ol.2018.7983] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 09/07/2017] [Indexed: 12/12/2022] Open
Abstract
Accumulating studies have demonstrated microRNAs (miRNAs/miRs) have an important role in multiple processes of human malignant tumor development and progression. Decreased expression of miR-125a-5p has been observed in several types of cancer, including gastric cancer (GC). However, the mechanism and exact function of miR-125a-5p in GC have not been largely elucidated. In the present study, reverse transcription-quantitative polymerase chain reaction indicated that the expression of miR-125a-5p was downregulated in GC tissues and cell lines compared with matched normal tissues (P<0.01) and normal gastric mucosa cell lines (P<0.01), respectively. Moreover, clinical pathological characteristics and Kaplan-Meier analysis indicated that a low expression of miR-125a-5p was not only associated with lymph metastasis, peritoneal dissemination and advanced tumor-node metastasis stage but also affected the prognosis of GC patients. Compared with miR-control-transfected GC cells, markedly decreased migration and invasion was observed in GC cells that overexpress miR-125a-5p. By contrast, increased metastasis and invasion were observed in miR-125a-5p-knocked down cells compared with the control. Furthermore, luciferase reporter assays indicated that breast cancer metastasis suppressor 1 (BRMS1) was a direct target of miR-125a-5p. Notably, a positive correlation between the levels of BRMS1 and miR-125a-5p in GC tissues was observed, and BRMS1 expression was indicated to be regulated by miR-125a-5p in GC cells. In conclusion, miR-125a-5p may act as a tumor suppressor by targeting the metastasis-inhibitory gene, BRMS1. The data suggesting that BRMS1 is a potential target gene of miR-125a-5p, may provide novel insight into miRNA regulation of human gene expression, and a useful target for gene therapy of GC.
Collapse
Affiliation(s)
- Yi Cao
- Department of General Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shengxing Tan
- Department of General Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yi Tu
- Department of Pathology, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Guoyang Zhang
- Department of General Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yi Liu
- Department of General Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Daojiang Li
- Department of General Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shan Xu
- Department of Pathology, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhibiao Le
- Department of General Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jianbo Xiong
- Department of General Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wenyu Zou
- Department of General Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Peitao Gong
- Department of General Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhengrong Li
- Department of General Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhigang Jie
- Department of General Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
7
|
Lee HJ, An HJ, Kim TH, Kim G, Kang H, Heo JH, Kwon AY, Kim S. Fascin expression is inversely correlated with breast cancer metastasis suppressor 1 and predicts a worse survival outcome in node-negative breast cancer patients. J Cancer 2017; 8:3122-3129. [PMID: 29158783 PMCID: PMC5665027 DOI: 10.7150/jca.22046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 08/29/2017] [Indexed: 12/03/2022] Open
Abstract
Background: Fascin is an actin-bundling protein that promotes cancer cell migration and invasion. By contrast, breast cancer metastasis suppressor 1 (BRMS1) inhibits cancer metastasis by targeting multiple steps of the metastatic cascade. We evaluated whether expression patterns of fascin and BRMS1 correlate with clinicopathological features and patient outcome. Methods: Immunohistochemistry for fascin and BRMS1 was performed using a tissue microarray constructed from 183 human breast cancer tissues. Fascin expression determined by the proportion of stained tumor cells (0: 0-5%, 1: 6-25%, 2: 26-50%, 3: 51-75%, or 4: >75%) and staining intensity (0: negative, 1: weak, 2: moderate, or 3: strong) were multiplied and defined as negative (0-3) or positive (4-12). BRMS1 expression was scored separately based on nuclear and cytoplasmic staining intensity (0: negative, 1: weak, 2: moderate, 3: strong). We obtained the BRMS1 H score by summing the nuclear and cytoplasmic scores and defined it as negative (0-2) or positive (3-6). Results: Expression of BRMS1 showed a significant inverse correlation with that of fascin. Fascin+ tumors were significantly associated with no lymph node metastasis, higher histological and higher nuclear grade, ER/PR/HER2 negativity, and triple-negative subtype (all ps < 0.05). These clinicopathological differences showed the same trend in a comparison of fascin-/BRMS1+ and fascin+/BRMS1- tumors. Negative or weak BRMS1 cytoplasmic expression was significantly associated with shorter disease-free survival (DFS; p = 0.043). Fascin positivity was significantly associated with shorter DFS (p = 0.005) and overall survival (p = 0.020) when analyses were confined to node-negative patients. Conclusions: This study confirms an inverse correlation between expression of fascin and expression of BRMS1 using a quite large cohort of human breast cancer tissues. Fascin alone or combined with BRMS1 was a worse prognostic marker, particularly in node-negative breast cancer patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sewha Kim
- Department of Pathology, CHA Bundang Medical Center, CHA University, 59 Yatap-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13496, Korea
| |
Collapse
|
8
|
Welch D, Manton C, Hurst D. Breast Cancer Metastasis Suppressor 1 (BRMS1): Robust Biological and Pathological Data, But Still Enigmatic Mechanism of Action. Adv Cancer Res 2016; 132:111-37. [PMID: 27613131 DOI: 10.1016/bs.acr.2016.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Metastasis requires coordinated expression of multiple genetic cassettes, often via epigenetic regulation of gene transcription. BRMS1 blocks metastasis, but not orthotopic tumor growth in multiple tumor types, presumably via SIN3 chromatin remodeling complexes. Although there is an abundance of strong data supporting BRMS1 as a metastasis suppressor, the mechanistic data directly connecting molecular pathways with inhibition of particular steps in metastasis are not well defined. In this review, the data for BRMS1-mediated metastasis suppression in multiple tumor types are discussed along with the steps in metastasis that are inhibited.
Collapse
|
9
|
Kodura MA, Souchelnytskyi S. Breast carcinoma metastasis suppressor gene 1 (BRMS1): update on its role as the suppressor of cancer metastases. Cancer Metastasis Rev 2015; 34:611-8. [DOI: 10.1007/s10555-015-9583-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
10
|
Xing WJ, Liao XH, Wang N, Zhao DW, Zheng L, Zheng DL, Dong J, Zhang TC. MRTF-A and STAT3 promote MDA-MB-231 cell migration via hypermethylating BRSM1. IUBMB Life 2015; 67:202-17. [PMID: 25854163 DOI: 10.1002/iub.1362] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 01/25/2015] [Accepted: 01/27/2015] [Indexed: 11/05/2022]
Abstract
Breast cancer is the leading cause of cancer death in women worldwide which is closely related to metastasis. But the exact molecular mechanism of metastasis is still not fully understood. We now report that both MRTF-A and STAT3 play important roles in migration of MDA-MB-231 breast cancer cells. Moreover, MRTF-A and STAT3 synergistically increased MDA-MB-231 cell migration by promoting the expression of migration markers urokinase-type plasminogen activator (uPA) and osteopontin (OPN) and inhibiting the expression of breast cancer metastasis suppressor 1 (BRMS1). Luciferase reporter assays demonstrated that MRTF-A and STAT3 do not affect transcription of the BRMS1 promoter. Instead, we identified a newly molecular mechanism by which MRTF-A and STAT3 synergistically controlled MDA-MB-231 cell migration by recruiting DNMT1 to hypermethylate the promoter of BRMS1 and thus affect the expression of BRMS1. Interestingly, physical interaction between MRTF-A and STAT3 synergistically promotes the transactivity of DNMT1 by binding to the GAS element within the DNMT1 promoter. Our data thus provide important and novel insights into the roles of MRTF-A and STAT3 in regulating MDA-MB-231 cell migration.
Collapse
Affiliation(s)
- Wen-Jing Xing
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
11
|
BRMS1L suppresses breast cancer metastasis by inducing epigenetic silence of FZD10. Nat Commun 2014; 5:5406. [PMID: 25406648 DOI: 10.1038/ncomms6406] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 09/29/2014] [Indexed: 12/16/2022] Open
Abstract
BRMS1L (breast cancer metastasis suppressor 1 like, BRMS1-like) is a component of Sin3A-histone deacetylase (HDAC) co-repressor complex that suppresses target gene transcription. Here we show that reduced BRMS1L in breast cancer tissues is associated with metastasis and poor patient survival. Functionally, BRMS1L inhibits breast cancer cells migration and invasion by inhibiting epithelial-mesenchymal transition. These effects are mediated by epigenetic silencing of FZD10, a receptor for Wnt signalling, through HDAC1 recruitment and histone H3K9 deacetylation at the promoter. Consequently, BRMS1L-induced FZD10 silencing inhibits aberrant activation of WNT3-FZD10-β-catenin signalling. Furthermore, BRMS1L is a target of miR-106b and miR-106b upregulation leads to BRMS1L reduction in breast cancer cells. RNA interference-mediated silencing of BRMS1L expression promotes metastasis of breast cancer xenografts in immunocompromised mice, whereas ectopic BRMS1L expression inhibits metastasis. Therefore, BRMS1L provides an epigenetic regulation of Wnt signalling in breast cancer cells and acts as a breast cancer metastasis suppressor.
Collapse
|