51
|
Vanneste M, Hanoux V, Bouakka M, Bonnamy PJ. Hyaluronate synthase-2 overexpression alters estrogen dependence and induces histone deacetylase inhibitor-like effects on ER-driven genes in MCF7 breast tumor cells. Mol Cell Endocrinol 2017; 444:48-58. [PMID: 28137613 DOI: 10.1016/j.mce.2017.01.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 01/26/2017] [Accepted: 01/26/2017] [Indexed: 10/20/2022]
Abstract
In breast carcinoma cells, high levels of hyaluronan (HA) and its CD44 receptor are frequently associated with alteration in estrogen signaling. We demonstrate that stable hyaluronate synthase 2 (HAS2) overexpression in estrogen receptor α (ERα) -positive MCF7 cells oppositely altered estrogen dependence of cell growth and its sensitivity towards antiestrogens. Albeit without effect on ERα expression and estradiol binding properties, HAS2 overexpression increased ERα Ser118 phosphorylation as well as transcriptional activity of estrogen in an ERE-luciferase reporter gene assay. However, HAS2 overexpression induced partial silencing of E2 driven-genes without affecting the magnitude of regulation by estradiol. This effect was associated with half-reduction in the activity of nuclear histone deacetylases (HDACs) through a post-translational mechanism likely consecutive to the enhanced expression of the histone acetyl-transferase EP300. In conclusion, increase in HA/CD44 interactions may contribute, through an HDAC inhibitor-like and ER-independent mechanism, to the silencing of estrogen-driven genes in breast carcinoma.
Collapse
Affiliation(s)
| | | | - Mohammed Bouakka
- Université Mohammed Premier, Faculté des Sciences, Laboratoire de Biochimie, Oujda, Morocco
| | | |
Collapse
|
52
|
Liu Y, Hou Y, Ma L, Sun C, Pan J, Yang Y, Zhou H, Zhang J. Regulation of semaphorin 4D expression and cell proliferation of ovarian cancer by ERalpha and ERbeta. ACTA ACUST UNITED AC 2017; 50:e6057. [PMID: 28225892 PMCID: PMC5333722 DOI: 10.1590/1414-431x20166057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 12/13/2016] [Indexed: 12/01/2022]
Abstract
Ovarian cancer is one of the most common malignancies in women. Semaphorin 4D (sema 4D) is involved in the progress of multiple cancers. In the presence of estrogen-like ligands, estrogen receptors (ERα and ERβ) participate in the progress of breast and ovarian cancers by transcriptional regulation. The aim of the study was to investigate the role of sema 4D and elucidate the regulatory pattern of ERα and ERβ on sema 4D expression in ovarian cancers. Sema 4D levels were up-regulated in ovarian cancer SKOV-3 cells. Patients with malignant ovarian cancers had significantly higher sema 4D levels than controls, suggesting an oncogene role of sema 4D in ovarian cancer. ERα expressions were up-regulated in SKOV-3 cells compared with normal ovarian IOSE80 epithelial cells. Conversely, down-regulation of ERβ was observed in SKOV-3 cells. Forced over-expression of ERα and ERβ in SKOV-3 cells was manipulated to establish ERα+ and ERβ+ SKOV-3 cell lines. Incubation of ERα+ SKOV-3 cells with ERs agonist 17β-estradiol (E2) significantly enhanced sema 4D expression and rate of cell proliferation. Incubated with E2, ERβ+ SKOV-3 cells showed lower sema 4D expression and cell proliferation. Blocking ERα and ERβ activities with ICI182-780 inhibitor, sema 4D expressions and cell proliferation of ERα+ and ERβ+ SKOV-3 cells were recovered to control levels. Taken together, the data showed that sema 4D expression was positively correlated with the progress of ovarian cancer. ERα positively regulated sema 4D expression and accelerated cell proliferation. ERβ negatively regulated sema 4D expression and inhibited cell multiplication.
Collapse
Affiliation(s)
- Y Liu
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Y Hou
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - L Ma
- Department of Reproduction and Genetics, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - C Sun
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - J Pan
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Y Yang
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - H Zhou
- Department of Gynecology, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - J Zhang
- Department of General Surgery, the Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
53
|
Dou XW, Liang YK, Lin HY, Wei XL, Zhang YQ, Bai JW, Chen CF, Chen M, Du CW, Li YC, Tian J, Man K, Zhang GJ. Notch3 Maintains Luminal Phenotype and Suppresses Tumorigenesis and Metastasis of Breast Cancer via Trans-Activating Estrogen Receptor-α. Theranostics 2017; 7:4041-4056. [PMID: 29109797 PMCID: PMC5667424 DOI: 10.7150/thno.19989] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 08/11/2017] [Indexed: 02/05/2023] Open
Abstract
The luminal A phenotype is the most common breast cancer subtype and is characterized by estrogen receptor α expression (ERα). Identification of the key regulator that governs the luminal phenotype of breast cancer will clarify the pathogenic mechanism and provide novel therapeutic strategies for this subtype of cancer. ERα signaling pathway sustains the epithelial phenotype and inhibits the epithelial-mesenchymal transition (EMT) of breast cancer. In this study, we demonstrate that Notch3 positively associates with ERα in both breast cancer cell lines and human breast cancer tissues. We found that overexpression of Notch3 intra-cellular domain, a Notch3 active form (N3ICD), in ERα negative breast cancer cells re-activated ERα, while knock-down of Notch3 reduced ERα transcript and proteins, with alteration of down-stream genes, suggesting its ability to regulate ERα. Mechanistically, our results show that Notch3 specifically binds to the CSL binding element of the ERα promoter and activates ERα expression. Moreover, Notch3 suppressed EMT, while suppression of Notch3 promoted EMT in cellular assay. Overexpressing N3ICD in triple-negative breast cancer suppressed tumorigenesis and metastasis in vivo. Conversely, depletion of Notch3 in luminal breast cancer promoted metastasis in vivo. Furthermore, Notch3 transcripts were significantly associated with prolonged relapse-free survival in breast cancer, in particular in ERα positive breast cancer patients. Our observations demonstrate that Notch3 governs the luminal phenotype via trans-activating ERα expression in breast cancer. These findings delineate the role of a Notch3/ERα axis in maintaining the luminal phenotype and inhibiting tumorigenesis and metastasis in breast cancer, providing a novel strategy to re-sensitize ERα negative or low-expressing breast cancers to hormone therapy.
Collapse
Affiliation(s)
- Xiao-Wei Dou
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
| | - Yuan-Ke Liang
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Hao-Yu Lin
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
- Department of Breast and Thyroid Surgery, the First Affiliated Hospital of SUMC
| | - Xiao-Long Wei
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
- Department of Pathology, the Cancer Hospital of Shantou University Medical College (SUMC), China
| | - Yong-Qu Zhang
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
| | - Jing-Wen Bai
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
| | - Chun-Fa Chen
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
| | - Min Chen
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
| | - Cai-Wen Du
- Department of Breast Medical Oncology, the Cancer Hospital of Shantou University Medical College (SUMC), China
| | - Yao-Chen Li
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
| | - Jie Tian
- Institute of Automation, Chinese Academy of Science, China
| | - Kwan Man
- Department of Surgery, Hong Kong University Li Ka-Tsing faculty of Medicine, Hong Kong, China
| | - Guo-Jun Zhang
- The Breast Center, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ChangJiang Scholar's Laboratory, the Cancer Hospital of Shantou University Medical College (SUMC), China
- ✉ Corresponding author: Guo-Jun Zhang, MD, PhD. Tel.: +86(754)88556826; E-mail:
| |
Collapse
|
54
|
Long-term exposure of MCF-7 breast cancer cells to ethanol stimulates oncogenic features. Int J Oncol 2016; 50:49-65. [PMID: 27959387 PMCID: PMC5182011 DOI: 10.3892/ijo.2016.3800] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/10/2016] [Indexed: 12/24/2022] Open
Abstract
Alcohol consumption is a risk factor for breast cancer. Little is known regarding the mechanism, although it is assumed that acetaldehyde or estrogen mediated pathways play a role. We previously showed that long-term exposure to 2.5 mM ethanol (blood alcohol ~0.012%) of MCF-12A, a human normal epithelial breast cell line, induced epithelial mesenchymal transition (EMT) and oncogenic transformation. In this study, we investigated in the human breast cancer cell line MCF-7, whether a similar exposure to ethanol at concentrations ranging up to peak blood levels in heavy drinkers would increase malignant progression. Short-term (1-week) incubation to ethanol at as low as 1-5 mM (corresponding to blood alcohol concentration of ~0.0048-0.024%) upregulated the stem cell related proteins Oct4 and Nanog, but they were reduced after exposure at 25 mM. Long-term (4-week) exposure to 25 mM ethanol upregulated the Oct4 and Nanog proteins, as well as the malignancy marker Ceacam6. DNA microarray analysis in cells exposed for 1 week showed upregulated expression of metallothionein genes, particularly MT1X. Long-term exposure upregulated expression of some malignancy related genes (STEAP4, SERPINA3, SAMD9, GDF15, KRT15, ITGB6, TP63, and PGR, as well as the CEACAM, interferon related, and HLA gene families). Some of these findings were validated by RT-PCR. A similar treatment also modulated numerous microRNAs (miRs) including one regulator of Oct4 as well as miRs involved in oncogenesis and/or malignancy, with only a few estrogen-induced miRs. Long-term 25 mM ethanol also induced a 5.6-fold upregulation of anchorage-independent growth, an indicator of malignant-like features. Exposure to acetaldehyde resulted in little or no effect comparable to that of ethanol. The previously shown alcohol induction of oncogenic transformation of normal breast cells is now complemented by the current results suggesting alcohol's potential involvement in malignant progression of breast cancer.
Collapse
|
55
|
Carthy JM, Stöter M, Bellomo C, Vanlandewijck M, Heldin A, Morén A, Kardassis D, Gahman TC, Shiau AK, Bickle M, Zerial M, Heldin CH, Moustakas A. Chemical regulators of epithelial plasticity reveal a nuclear receptor pathway controlling myofibroblast differentiation. Sci Rep 2016; 6:29868. [PMID: 27430378 PMCID: PMC4949434 DOI: 10.1038/srep29868] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 06/27/2016] [Indexed: 12/23/2022] Open
Abstract
Plasticity in epithelial tissues relates to processes of embryonic development, tissue fibrosis and cancer progression. Pharmacological modulation of epithelial transitions during disease progression may thus be clinically useful. Using human keratinocytes and a robotic high-content imaging platform, we screened for chemical compounds that reverse transforming growth factor β (TGF-β)-induced epithelial-mesenchymal transition. In addition to TGF-β receptor kinase inhibitors, we identified small molecule epithelial plasticity modulators including a naturally occurring hydroxysterol agonist of the liver X receptors (LXRs), members of the nuclear receptor transcription factor family. Endogenous and synthetic LXR agonists tested in diverse cell models blocked α-smooth muscle actin expression, myofibroblast differentiation and function. Agonist-dependent LXR activity or LXR overexpression in the absence of ligand counteracted TGF-β-mediated myofibroblast terminal differentiation and collagen contraction. The protective effect of LXR agonists against TGF-β-induced pro-fibrotic activity raises the possibility that anti-lipidogenic therapy may be relevant in fibrotic disorders and advanced cancer.
Collapse
Affiliation(s)
- Jon M Carthy
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, Biomedical Center, SE-751 24 Uppsala, Sweden
| | - Martin Stöter
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Claudia Bellomo
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, Biomedical Center, SE-751 24 Uppsala, Sweden.,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Box 582, Biomedical Center, SE-751 23 Uppsala, Sweden
| | - Michael Vanlandewijck
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, Biomedical Center, SE-751 24 Uppsala, Sweden
| | - Angelos Heldin
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, Biomedical Center, SE-751 24 Uppsala, Sweden
| | - Anita Morén
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, Biomedical Center, SE-751 24 Uppsala, Sweden
| | - Dimitris Kardassis
- Department of Biochemistry, University of Crete Medical School, 71003 Heraklion, Crete, Greece
| | - Timothy C Gahman
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | - Andrew K Shiau
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA
| | - Marc Bickle
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Marino Zerial
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Carl-Henrik Heldin
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, Biomedical Center, SE-751 24 Uppsala, Sweden
| | - Aristidis Moustakas
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, Biomedical Center, SE-751 24 Uppsala, Sweden.,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Box 582, Biomedical Center, SE-751 23 Uppsala, Sweden
| |
Collapse
|
56
|
EMT in Breast Carcinoma-A Review. J Clin Med 2016; 5:jcm5070065. [PMID: 27429011 PMCID: PMC4961996 DOI: 10.3390/jcm5070065] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 07/01/2016] [Accepted: 07/06/2016] [Indexed: 02/07/2023] Open
Abstract
The epithelial to mesenchymal transition (EMT) is a cellular program that is involved in embryonic development; wound healing, but also in tumorigenesis. Breast carcinoma (BC) is the most common cancer in women worldwide, and the majority of deaths (90%) are caused by invasion and metastasis. The EMT plays an important role in invasion and subsequent metastasis. Several distinct biological events integrate a cascade that leads not only to a change from an epithelial to mesenchymal phenotype, but allows for detachment, migration, invasion and ultimately, colonization of a second site. Understanding the biological intricacies of the EMT may provide important insights that lead to the development of therapeutic targets in pre-invasive and invasive breast cancer, and could be used as biomarkers identifying tumor subsets with greater chances of recurrence, metastasis and therapeutic resistance leading to death.
Collapse
|
57
|
Choe M, Brusgard JL, Chumsri S, Bhandary L, Zhao XF, Lu S, Goloubeva OG, Polster BM, Fiskum GM, Girnun GD, Kim MS, Passaniti A. The RUNX2 Transcription Factor Negatively Regulates SIRT6 Expression to Alter Glucose Metabolism in Breast Cancer Cells. J Cell Biochem 2016; 116:2210-26. [PMID: 25808624 DOI: 10.1002/jcb.25171] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 03/20/2015] [Indexed: 12/21/2022]
Abstract
Activation of genes promoting aerobic glycolysis and suppression of mitochondrial oxidative phosphorylation is one of the hallmarks of cancer. The RUNX2 transcription factor mediates breast cancer (BC) metastasis to bone and is regulated by glucose availability. But, the mechanisms by which it regulates glucose metabolism and promotes an oncogenic phenotype are not known. RUNX2 expression in luminal BC cells correlated with lower estrogen receptor-α (ERα) levels, anchorage-independent growth, expression of glycolytic genes, increased glucose uptake, and sensitivity to glucose starvation, but not to inhibitors of oxidative phosphorylation. Conversely, RUNX2 knockdown in triple-negative BC cells inhibited mammosphere formation and glucose dependence. RUNX2 knockdown resulted in lower LDHA, HK2, and GLUT1 glycolytic gene expression, but upregulation of pyruvate dehydrogenase-A1 (PDHA1) mRNA and enzymatic activity, which was consistent with lower glycolytic potential. The NAD-dependent histone deacetylase, SIRT6, a known tumor suppressor, was a critical regulator of these RUNX2-mediated metabolic changes. RUNX2 expression resulted in elevated pAkt, HK2, and PDHK1 glycolytic protein levels that were reduced by ectopic expression of SIRT6. RUNX2 also repressed mitochondrial oxygen consumption rates (OCR), a measure of oxidative phosphorylation (respiration). Overexpression of SIRT6 increased respiration in RUNX2-positive cells, but knockdown of SIRT6 in cells expressing low RUNX2 decreased respiration. RUNX2 repressed SIRT6 expression at both the transcriptional and post-translational levels and endogenous SIRT6 expression was lower in malignant BC tissues or cell lines that expressed high levels of RUNX2. These results support a hypothesis whereby RUNX2-mediated repression of the SIRT6 tumor suppressor regulates metabolic pathways that promote BC progression.
Collapse
Affiliation(s)
- Moran Choe
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland.,Laboratory of Genitourinary Cancer Pathogenesis, NCI, Building 36/Room 1130, 37 Convent Drive, Bethesda, Maryland, 20814
| | - Jessica L Brusgard
- Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Saranya Chumsri
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Lekhana Bhandary
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Xianfeng Frank Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Pathology and the VA San Diego Healthcare System, University of California, San Diego, California, 92161
| | - Song Lu
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Olga G Goloubeva
- Department of Epidemiology & Public Health, The Marlene & Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Brian M Polster
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Gary M Fiskum
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Geoffrey D Girnun
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Pathology, Stony Brook University Medical Center, Stony Brook, New York, 11794
| | - Myoung Sook Kim
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Antonino Passaniti
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland.,Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland.,The Veteran's Health Administration Research & Development Service, Marlene & Stewart Greenebaum Cancer Center, University of Maryland, Baltimore, Maryland
| |
Collapse
|
58
|
Huo L, Wang Y, Gong Y, Krishnamurthy S, Wang J, Diao L, Liu CG, Liu X, Lin F, Symmans WF, Wei W, Zhang X, Sun L, Alvarez RH, Ueno NT, Fouad TM, Harano K, Debeb BG, Wu Y, Reuben J, Cristofanilli M, Zuo Z. MicroRNA expression profiling identifies decreased expression of miR-205 in inflammatory breast cancer. Mod Pathol 2016; 29:330-46. [PMID: 26916073 PMCID: PMC11793840 DOI: 10.1038/modpathol.2016.38] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/09/2016] [Accepted: 01/09/2016] [Indexed: 02/07/2023]
Abstract
Inflammatory breast cancer is the most aggressive form of breast cancer. Identifying new biomarkers to be used as therapeutic targets is in urgent need. Messenger RNA expression profiling studies have indicated that inflammatory breast cancer is a transcriptionally heterogeneous disease, and specific molecular targets for inflammatory breast cancer have not been well established. We performed microRNA expression profiling in inflammatory breast cancer in comparison with locally advanced noninflammatory breast cancer in this study. Although many microRNAs were differentially expressed between normal breast tissue and tumor tissue, most of them did not show differential expression between inflammatory and noninflammatory tumor samples. However, by microarray analysis, quantitative reverse transcription PCR, and in situ hybridization, we showed that microRNA-205 expression was decreased not only in tumor compared with normal breast tissue, but also in inflammatory breast cancer compared with noninflammatory breast cancer. Lower expression of microRNA-205 correlated with worse distant metastasis-free survival and overall survival in our cohort. A small-scale immunohistochemistry analysis showed coexistence of decreased microRNA-205 expression and decreased E-cadherin expression in some ductal tumors. MicroRNA-205 may serve as a therapeutic target in advanced breast cancer including inflammatory breast cancer.
Collapse
Affiliation(s)
- Lei Huo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Yan Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Yun Gong
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Savitri Krishnamurthy
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Lixia Diao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Chang-Gong Liu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Xiuping Liu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Feng Lin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - William F. Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Wei Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Xinna Zhang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Li Sun
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Ricardo H. Alvarez
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Naoto T. Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Tamer M. Fouad
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Kenichi Harano
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Bisrat G. Debeb
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - Yun Wu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | - James Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| | | | - Zhuang Zuo
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United State
| |
Collapse
|
59
|
Gelfand R, Vernet D, Bruhn K, Vadgama J, Gonzalez-Cadavid NF. Long-term exposure of MCF-12A normal human breast epithelial cells to ethanol induces epithelial mesenchymal transition and oncogenic features. Int J Oncol 2016; 48:2399-414. [PMID: 27035792 PMCID: PMC4864041 DOI: 10.3892/ijo.2016.3461] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 02/24/2016] [Indexed: 12/12/2022] Open
Abstract
Alcoholism is associated with breast cancer incidence and progression, and moderate chronic consumption of ethanol is a risk factor. The mechanisms involved in alcohol's oncogenic effects are unknown, but it has been speculated that they may be mediated by acetaldehyde. We used the immortalized normal human epithelial breast cell line MCF-12A to determine whether short- or long-term exposure to ethanol or to acetaldehyde, using in vivo compatible ethanol concentrations, induces their oncogenic transformation and/or the acquisition of epithelial mesenchymal transition (EMT). Cultures of MCF-12A cells were incubated with 25 mM ethanol or 2.5 mM acetaldehyde for 1 week, or with lower concentrations (1.0–2.5 mM for ethanol, 1.0 mM for acetaldehyde) for 4 weeks. In the 4-week incubation, cells were also tested for anchorage-independence, including isolation of soft agar selected cells (SASC) from the 2.5 mM ethanol incubations. Cells were analyzed by immunocytofluorescence, flow cytometry, western blotting, DNA microarrays, RT/PCR, and assays for miRs. We found that short-term exposure to ethanol, but not, in general, to acetaldehyde, was associated with transcriptional upregulation of the metallothionein family genes, alcohol metabolism genes, and genes suggesting the initiation of EMT, but without related phenotypic changes. Long-term exposure to the lower concentrations of ethanol or acetaldehyde induced frank EMT changes in the monolayer cultures and in SASC as demonstrated by changes in cellular phenotype, mRNA expression, and microRNA expression. This suggests that low concentrations of ethanol, with little or no mediation by acetaldehyde, induce EMT and some traits of oncogenic transformation such as anchorage-independence in normal breast epithelial cells.
Collapse
Affiliation(s)
- Robert Gelfand
- Department of Medicine, Charles Drew University (CDU), Los Angeles, CA, USA
| | - Dolores Vernet
- Department of Medicine, Charles Drew University (CDU), Los Angeles, CA, USA
| | - Kevin Bruhn
- Department of Surgery, Los Angeles Biomedical Research Institute (LABioMed) at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jaydutt Vadgama
- Department of Medicine, Charles Drew University (CDU), Los Angeles, CA, USA
| | | |
Collapse
|
60
|
Regulation of epithelial-mesenchymal transition in endometrial cancer: connecting PI3K, estrogen signaling, and microRNAs. Clin Transl Oncol 2016; 18:1056-1061. [PMID: 26856598 DOI: 10.1007/s12094-016-1492-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 01/27/2016] [Indexed: 02/07/2023]
Abstract
Endometrial cancer (EC) prognosis is dependent on many factors such as time of diagnosis, histological type, and degree of invasion. Type I EC has a more favorable prognosis as it is less prone to myometrial invasion, which is believed to be the first step in the metastatic cascade. Type II EC displays a more aggressive and motile phenotype, and therefore has a poorer prognosis. Recent work suggests that despite the epithelial nature of Type I and Type II endometrial tumors, both are capable of undergoing an epithelial-mesenchymal transition (EMT), which may facilitate myometrial invasion and metastasis. Activation of the PI3K/Akt pathway has been shown to contribute to EMT through the upregulation of EMT-associated factors. Recent research has also linked estrogen signaling and microRNAs to the regulatory mechanisms that drive EMT in EC. Understanding the intricate relationships between these pathways will provide a better understanding of metastatic progression in EC.
Collapse
|
61
|
Voutsadakis IA. Epithelial-Mesenchymal Transition (EMT) and Regulation of EMT Factors by Steroid Nuclear Receptors in Breast Cancer: A Review and in Silico Investigation. J Clin Med 2016; 5:E11. [PMID: 26797644 PMCID: PMC4730136 DOI: 10.3390/jcm5010011] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 12/23/2015] [Accepted: 12/30/2015] [Indexed: 12/20/2022] Open
Abstract
Steroid Nuclear Receptors (SNRs) are transcription factors of the nuclear receptor super-family. Estrogen Receptor (ERα) is the best-studied and has a seminal role in the clinic both as a prognostic marker but also as a predictor of response to anti-estrogenic therapies. Progesterone Receptor (PR) is also used in the clinic but with a more debatable prognostic role and the role of the four other SNRs, ERβ, Androgen Receptor (AR), Glucocorticoid Receptor (GR) and Mineralocorticoid Receptor (MR), is starting only to be appreciated. ERα, but also to a certain degree the other SNRs, have been reported to be involved in virtually every cancer-enabling process, both promoting and impeding carcinogenesis. Epithelial-Mesenchymal Transition (EMT) and the reverse Mesenchymal Epithelial Transition (MET) are such carcinogenesis-enabling processes with important roles in invasion and metastasis initiation but also establishment of tumor in the metastatic site. EMT is governed by several signal transduction pathways culminating in core transcription factors of the process, such as Snail, Slug, ZEB1 and ZEB2, and Twist, among others. This paper will discuss direct regulation of these core transcription factors by SNRs in breast cancer. Interrogation of publicly available databases for binding sites of SNRs on promoters of core EMT factors will also be included in an attempt to fill gaps where other experimental data are not available.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Division of Medical Oncology, Department of Internal Medicine, Sault Area Hospital, Sault Ste Marie, ON P6B 0A8, Canada.
- Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, QC P3E 2C6, Canada.
| |
Collapse
|
62
|
Lo PK, Wolfson B, Zhou X, Duru N, Gernapudi R, Zhou Q. Noncoding RNAs in breast cancer. Brief Funct Genomics 2015; 15:200-21. [PMID: 26685283 DOI: 10.1093/bfgp/elv055] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The mammalian transcriptome has recently been revealed to encompass a large number of noncoding RNAs (ncRNAs) that play a variety of important regulatory roles in gene expression and other biological processes. MicroRNAs (miRNAs), the best studied of the short noncoding RNAs (sncRNAs), have been extensively characterized with regard to their biogenesis, function and importance in tumorigenesis. Another class of sncRNAs called piwi-interacting RNAs (piRNAs) has also gained attention recently in cancer research owing to their critical role in stem cell regulation. Long noncoding RNAs (lncRNAs) of >200 nucleotides in length have recently emerged as key regulators of developmental processes, including mammary gland development. lncRNA dysregulation has also been implicated in the development of various cancers, including breast cancer. In this review, we describe and discuss the roles of sncRNAs (including miRNAs and piRNAs) and lncRNAs in the initiation and progression of breast tumorigenesis, with a focus on outlining the molecular mechanisms of oncogenic and tumor-suppressor ncRNAs. Moreover, the current and potential future applications of ncRNAs to clinical breast cancer research are also discussed, with an emphasis on ncRNA-based diagnosis, prognosis and future therapeutics.
Collapse
|
63
|
Klinge CM. miRNAs regulated by estrogens, tamoxifen, and endocrine disruptors and their downstream gene targets. Mol Cell Endocrinol 2015; 418 Pt 3:273-97. [PMID: 25659536 PMCID: PMC4523495 DOI: 10.1016/j.mce.2015.01.035] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 01/22/2015] [Accepted: 01/23/2015] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNAs) are short (22 nucleotides), single-stranded, non-coding RNAs that form complimentary base-pairs with the 3' untranslated region of target mRNAs within the RNA-induced silencing complex (RISC) and block translation and/or stimulate mRNA transcript degradation. The non-coding miRBase (release 21, June 2014) reports that human genome contains ∼ 2588 mature miRNAs which regulate ∼ 60% of human protein-coding mRNAs. Dysregulation of miRNA expression has been implicated in estrogen-related diseases including breast cancer and endometrial cancer. The mechanism for estrogen regulation of miRNA expression and the role of estrogen-regulated miRNAs in normal homeostasis, reproduction, lactation, and in cancer is an area of great research and clinical interest. Estrogens regulate miRNA transcription through estrogen receptors α and β in a tissue-specific and cell-dependent manner. This review focuses primarily on the regulation of miRNA expression by ligand-activated ERs and their bona fide gene targets and includes miRNA regulation by tamoxifen and endocrine disrupting chemicals (EDCs) in breast cancer and cell lines.
Collapse
Affiliation(s)
- Carolyn M Klinge
- Department of Biochemistry & Molecular Biology, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, KY 40292, USA.
| |
Collapse
|
64
|
McFall T, Patki M, Rosati R, Ratnam M. Role of the short isoform of the progesterone receptor in breast cancer cell invasiveness at estrogen and progesterone levels in the pre- and post-menopausal ranges. Oncotarget 2015; 6:33146-64. [PMID: 26356672 PMCID: PMC4741755 DOI: 10.18632/oncotarget.5082] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 08/14/2015] [Indexed: 12/20/2022] Open
Abstract
Overexpression of the progesterone receptor (PR) isoform A (PR-A) is a negative prognosticator for estrogen receptor (ER)-positive breast cancer but in vitro studies have implicated PR-B in progestin-induced invasiveness. As estrogen is known to suppress invasiveness and tumor progression and as the in vitro studies were conducted in models that either lacked ER or excluded estrogen, we examined the role of PR isoforms in the context of estrogen signaling. Estrogen (< 0.01nM) strongly suppressed invasiveness in various ER+ model cell lines. At low (< 1nM) concentrations, progestins completely abrogated inhibition of invasiveness by estrogen. It was only in a higher (5 nM - 50 nM) concentration range that progestins induced invasiveness in the absence of estrogen. The ability of low dose progestins to rescue invasiveness from estrogen regulation was exclusively mediated by PR-A, whereas PR-B mediated the estrogen-independent component of progestin-induced invasiveness. Overexpression of PR-A lowered the progestin concentration needed to completely rescue invasiveness. Among estrogen-regulated genes, progestin/PR-A counter-regulated a distinctive subset, including breast tumor progression genes (e.g., HES1, PRKCH, ELF5, TM4SF1), leading to invasiveness. In this manner, at relatively low hormone concentrations (corresponding to follicular stage and post-menopausal breast tissue or plasma levels), progesterone influences breast cancer cell invasiveness by rescuing it from estrogen regulation via PR-A, whereas at higher concentrations the hormone also induces invasiveness independent of estrogen signaling, through PR-B. The findings point to a direct functional link between PR-A and progression of luminal breast cancer in the context of the entire range of pre- and post-menopausal plasma and breast tissue hormone levels.
Collapse
Affiliation(s)
- Thomas McFall
- Barbara Ann Karmanos Cancer Institute and Department of Oncology, Wayne State University, Detroit, MI, USA
| | - Mugdha Patki
- Barbara Ann Karmanos Cancer Institute and Department of Oncology, Wayne State University, Detroit, MI, USA
| | - Rayna Rosati
- Barbara Ann Karmanos Cancer Institute and Department of Oncology, Wayne State University, Detroit, MI, USA
| | - Manohar Ratnam
- Barbara Ann Karmanos Cancer Institute and Department of Oncology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
65
|
Di Leva G, Cheung DG, Croce CM. miRNA clusters as therapeutic targets for hormone-resistant breast cancer. Expert Rev Endocrinol Metab 2015; 10:607-617. [PMID: 27721895 PMCID: PMC5053393 DOI: 10.1586/17446651.2015.1099430] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
MicroRNAs are small non coding RNAs that typically inhibit the translation and stability of messenger RNAs, controlling genes involved in cellular processes such as inflammation, cell cycle regulation, stress response, differentiation, apoptosis, and migration. Not surprisingly, microRNAs are also aberrantly expressed in cancer and promote tumorigenesis by disrupting these vital cellular functions. In this review, we first broadly summarize the role of microRNAs in breast cancer and Estrogen Receptor alpha signaling. Then we focus on what is currently known about the role of microRNAs in anti-hormonal therapy or resistance to endocrine agents. Specifically, we will discuss key miRNAs involved in tamoxifen (miR-221/222, 181, 101, 519a, 301, 375, 342, 451, and the let-7 family), fulvestrant (miR-221/222, miR-200 family), and aromatase inhibitor (miR-128 and the let-7 family) resistance.
Collapse
Affiliation(s)
- Gianpiero Di Leva
- Department of Molecular Virology Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Douglas G Cheung
- Department of Molecular Virology Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Carlo M Croce
- Department of Molecular Virology Immunology and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| |
Collapse
|
66
|
Wei XL, Dou XW, Bai JW, Luo XR, Qiu SQ, Xi DD, Huang WH, Du CW, Man K, Zhang GJ. ERα inhibits epithelial-mesenchymal transition by suppressing Bmi1 in breast cancer. Oncotarget 2015; 6:21704-17. [PMID: 26023734 PMCID: PMC4673297 DOI: 10.18632/oncotarget.3966] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 04/30/2015] [Indexed: 02/05/2023] Open
Abstract
In human breast cancer, estrogen receptor-α (ERα) suppresses epithelial-mesenchymal transition (EMT) and stemness, two crucial parameters for tumor metastasis; however, the underlying mechanism by which ERα regulates these two processes remains largely unknown. Bmi1, the polycomb group protein B lymphoma Mo-MLV insertion region 1 homolog, regulates EMT transition, maintains the self-renewal capacity of stem cells, and is frequently overexpressed in human cancers. In the present study, ERα upregulated the expression of the epithelial marker, E-cadherin, in breast cancer cells through the transcriptional down-regulation of Bmi1. Furthermore, ERα overexpression suppressed the migration, invasion, and EMT of breast cancer cells. Notably, overexpression of ERα significantly decreased the CD44high/CD24low cell population and inhibited the capacity for mammosphere formation in ERα-negative breast cancer cells. In addition, overexpression of Bmi1 attenuated the ERα-mediated suppression of EMT and cell stemness. Immunohistochemistry revealed an inverse association of ERα and Bmi1 expression in human breast cancer tissue. Taken together, our findings suggest that ERα inhibits EMT and stemness through the downregulation of Bmi1.
Collapse
Affiliation(s)
- Xiao-Long Wei
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou 515031, China
- Changjiang Scholar's Laboratory and Cancer Research Center, Shantou University Medical College, Shantou 515031, China
| | - Xiao-Wei Dou
- Changjiang Scholar's Laboratory and Cancer Research Center, Shantou University Medical College, Shantou 515031, China
| | - Jing-Wen Bai
- Changjiang Scholar's Laboratory and Cancer Research Center, Shantou University Medical College, Shantou 515031, China
| | - Xiang-Rong Luo
- Changjiang Scholar's Laboratory and Cancer Research Center, Shantou University Medical College, Shantou 515031, China
| | - Si-Qi Qiu
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Di-Di Xi
- Changjiang Scholar's Laboratory and Cancer Research Center, Shantou University Medical College, Shantou 515031, China
| | - Wen-He Huang
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Cai-Wen Du
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Kwan Man
- Department of Surgery and Transplantation, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong 999077, China
| | - Guo-Jun Zhang
- Changjiang Scholar's Laboratory and Cancer Research Center, Shantou University Medical College, Shantou 515031, China
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| |
Collapse
|
67
|
Czarnecka AM, Kornakiewicz A, Lian F, Szczylik C. Future perspectives for mTOR inhibitors in renal cell cancer treatment. Future Oncol 2015; 11:801-17. [DOI: 10.2217/fon.14.303] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
ABSTRACT Everolimus is a mTOR inhibitor that demonstrates antitumor and antiangiogenic activities. In a randomized Phase III trial, patients with metastatic renal cell carcinoma who progressed on sunitinib/sorafenib were treated with everolimus and showed significant improvement in progression-free survival compared with best supportive care. Novel approaches in treatment are expected to ensure less toxic therapies and increase efficacy of everolimus. To provide a new perspective for mTOR inhibitor research and therapy, we discuss renal cell carcinoma cancer stem cells as a potential target for mTOR inhibitors and present new concepts on emerging antiangiogenic therapies. Finally, we point why systems biology approach with reverse molecular engineering may also contribute to the field of drug discovery in renal cell carcinoma.
Collapse
Affiliation(s)
- Anna M Czarnecka
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| | - Anna Kornakiewicz
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Fei Lian
- Emory School of Medicine Atlanta, GA 30322, USA
| | - Cezary Szczylik
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| |
Collapse
|
68
|
Murray JI, West NR, Murphy LC, Watson PH. Intratumoural inflammation and endocrine resistance in breast cancer. Endocr Relat Cancer 2015; 22:R51-67. [PMID: 25404688 DOI: 10.1530/erc-14-0096] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It is becoming clear that inflammation-associated mechanisms can affect progression of breast cancer and modulate responses to treatment. Estrogen receptor alpha (ERα (ESR1)) is the principal biomarker and therapeutic target for endocrine therapies in breast cancer. Over 70% of patients are ESR1-positive at diagnosis and are candidates for endocrine therapy. However, ESR1-positive tumours can become resistant to endocrine therapy. Multiple mechanisms of endocrine resistance have been proposed, including suppression of ESR1. This review discusses the relationship between intratumoural inflammation and endocrine resistance with a particular focus on inflammation-mediated suppression of ESR1.
Collapse
Affiliation(s)
- Jill I Murray
- Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada
| | - Nathan R West
- Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada
| | - Leigh C Murphy
- Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada
| | - Peter H Watson
- Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada Deeley Research CentreBritish Columbia Cancer Agency, 2410 Lee Avenue, Victoria, British Columbia, Canada V8R 6V5Translational Gastroenterology UnitNuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UKDepartment of Biochemistry and Medical Genetics and the Manitoba Institute of Cell BiologyUniversity of Manitoba and CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, CanadaDepartment of Biochemistry and MicrobiologyUniversity of Victoria, Victoria, British Columbia, CanadaDepartment of Pathology and Laboratory MedicineUniversity of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
69
|
Patki M, Salazar MD, Trumbly R, Ratnam M. Differential effects of estrogen-dependent transactivation vs. transrepression by the estrogen receptor on invasiveness of HER2 overexpressing breast cancer cells. Biochem Biophys Res Commun 2015; 457:404-11. [PMID: 25582774 DOI: 10.1016/j.bbrc.2015.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 01/05/2015] [Indexed: 02/03/2023]
Abstract
Estrogen (E2) supports breast cancer cell growth but suppresses invasiveness and both actions are antagonized by anti-estrogens. As a consequence, anti-estrogen treatment may increase the invasive potential of estrogen receptor (ER)+ tumor cell sub-populations that are endocrine resistant due to HER2 amplification. Either transactivation or transrepression by E2/ER could lead to both up- and down-regulation of many genes. Inhibition of the transactivation function of ER is adequate to inhibit E2-dependent growth. However, the impact of inhibiting E2-dependent transactivation vs. transrepression by ER on regulation of invasiveness by E2 is less clear. Here we dissect the roles of ER-mediated transactivation and transrepression in the regulation of invasiveness of ER+/HER2+ breast cancer cells by E2. Knocking down the general ER co-activators CBP and p300 prevented activation by E2 of its classical target genes but did not interfere with the ability of E2 to repress its direct target genes known to support invasiveness and tumor progression; there was also no effect on invasiveness or the ability of E2 to regulate invasiveness. On the other hand, overexpression of a co-repressor binding site mutant of ER (L372R) prevented E2-dependent transrepression but not transactivation. The mutant ER abrogated the ability of E2 to suppress invasiveness. E2 can partially down-regulate HER2 but knocking down HER2 below E2-regulated levels did not affect invasiveness or the ability of E2 to regulate invasiveness, although it did inhibit growth. Therefore, in ER+/HER2+ cells, the E2-dependent transrepression by ER rather than its transactivation function is critical for regulation of invasiveness and this is independent of HER2 regulation by E2. The findings suggest that selective inhibitors of transactivation by ER may be more beneficial in reducing tumor progression than conventional anti-estrogens that also antagonize E2-dependent transrepression.
Collapse
Affiliation(s)
- Mugdha Patki
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, 4100 John R., Detroit, MI 48201, USA; Department of Biochemistry and Cancer Biology, University Medical Center, Toledo, OH 43614, USA
| | - Marcela d'alincourt Salazar
- Department of Biochemistry and Cancer Biology, University Medical Center, Toledo, OH 43614, USA; Division of Translational Research, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Robert Trumbly
- Department of Biochemistry and Cancer Biology, University Medical Center, Toledo, OH 43614, USA
| | - Manohar Ratnam
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, 4100 John R., Detroit, MI 48201, USA.
| |
Collapse
|
70
|
Shashova EE, Lyupina YV, Glushchenko SA, Slonimskaya EM, Savenkova OV, Kulikov AM, Gornostaev NG, Kondakova IV, Sharova NP. Proteasome functioning in breast cancer: connection with clinical-pathological factors. PLoS One 2014; 9:e109933. [PMID: 25329802 PMCID: PMC4201529 DOI: 10.1371/journal.pone.0109933] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 08/12/2014] [Indexed: 01/25/2023] Open
Abstract
Breast cancer is one of four oncology diseases that are most widespread in the world. Moreover, breast cancer is one of leading causes of cancer-related deaths in female population within economically developed regions of the world. So far, detection of new mechanisms of breast cancer development is very important for discovery of novel areas in which therapy approaches may be elaborated. The objective of the present study is to investigate involvement of proteasomes, which cleave up to 90% of cellular proteins and regulate numerous cellular processes, in mechanisms of breast cancer development. Proteasome characteristics in 106 patient breast carcinomas and adjacent tissues, as well as relationships of detected proteasome parameters with clinical-pathological factors, were investigated. Proteasome chymotrypsin-like activity was evaluated by hydrolysis of fluorogenic peptide Suc-LLVY-AMC. The expression of proteasome subunits was studied by Western-blotting and immunohistochemistry. The wide range of chymotrypsin-like activity in tumors was detected. Activity in tumors was higher if compared to adjacent tissues in 76 from 106 patients. Multiple analysis of generalized linear models discovered that in estrogen α-receptor absence, tumor growth was connected with the enhanced expression of proteasome immune subunit LMP2 and proteasome activator PA700 in tumor (at 95% confidence interval). Besides, by this analysis we detected some phenomena in adjacent tissue, which are important for tumor growth and progression of lymph node metastasis in estrogen α-receptor absence. These phenomena are related to the enhanced expression of activator PA700 and immune subunit LMP7. Thus, breast cancer development is connected with functioning of immune proteasome forms and activator PA700 in patients without estrogen α-receptors in tumor cells. These results could indicate a field for search of new therapy approaches for this category of patients, which has the worst prognosis of health recovery.
Collapse
Affiliation(s)
- Elena E. Shashova
- Department of Experimental Oncology, Cancer Research Institute of Siberian Branch of Russian Academy of Medical Sciences, Tomsk, Russia
| | - Yulia V. Lyupina
- Department of Biochemistry of Ontogenesis Processes, NK Koltsov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| | - Svetlana A. Glushchenko
- Department of Pathological Anatomy and Cytology, Cancer Research Institute of Siberian Branch of Russian Academy of Medical Sciences, Tomsk, Russia
| | - Elena M. Slonimskaya
- Department of General Oncology, Cancer Research Institute of Siberian Branch of Russian Academy of Medical Sciences, Tomsk, Russia
- Department of Oncology, Siberian State Medical University, Tomsk, Russia
| | - Olga V. Savenkova
- Department of Pathological Anatomy and Cytology, Cancer Research Institute of Siberian Branch of Russian Academy of Medical Sciences, Tomsk, Russia
| | - Alexey M. Kulikov
- Department of Evolutionary and Developmental Genetics, NK Koltsov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| | - Nikolay G. Gornostaev
- Department of Evolutionary and Developmental Genetics, NK Koltsov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| | - Irina V. Kondakova
- Department of Experimental Oncology, Cancer Research Institute of Siberian Branch of Russian Academy of Medical Sciences, Tomsk, Russia
| | - Natalia P. Sharova
- Department of Biochemistry of Ontogenesis Processes, NK Koltsov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
71
|
McCubrey JA, Davis NM, Abrams SL, Montalto G, Cervello M, Libra M, Nicoletti F, D'Assoro AB, Cocco L, Martelli AM, Steelman LS. Targeting breast cancer initiating cells: advances in breast cancer research and therapy. Adv Biol Regul 2014; 56:81-107. [PMID: 24913694 DOI: 10.1016/j.jbior.2014.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 05/06/2014] [Indexed: 06/03/2023]
Abstract
Over the past 10 years there have been significant advances in our understanding of breast cancer and the important roles that breast cancer initiating cells (CICs) play in the development and resistance of breast cancer. Breast CICs endowed with self-renewing and tumor-initiating capacities are believed to be responsible for the relapses which often occur after various breast cancer therapies. In this review, we will summarize some of the key developments in breast CICs which will include discussion of some of the key genes implicated: estrogen receptor (ER), HER2, BRCA1, TP53, PIK3CA, RB, P16INK1 and various miRs as well some drugs which are showing promise in targeting CICs. In addition, the concept of combined therapies will be discussed. Basic and clinical research is resulting in novel approaches to improve breast cancer therapy by targeting the breast CICs.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Brody Building 5N98C, Greenville, NC 27858, USA.
| | - Nicole M Davis
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Brody Building 5N98C, Greenville, NC 27858, USA
| | - Stephen L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Brody Building 5N98C, Greenville, NC 27858, USA
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Massimo Libra
- Department of Bio-Medical Sciences, University of Catania, Catania, Italy
| | | | - Antonino B D'Assoro
- Department of Medical Oncology, Mayo Clinic Cancer Center, Rochester, MN, USA
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Brody Building 5N98C, Greenville, NC 27858, USA
| |
Collapse
|
72
|
Li Y, Li W, Ying Z, Tian H, Zhu X, Li J, Li M. Metastatic heterogeneity of breast cancer cells is associated with expression of a heterogeneous TGFβ-activating miR424-503 gene cluster. Cancer Res 2014; 74:6107-18. [PMID: 25164015 DOI: 10.1158/0008-5472.can-14-0389] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
TGFβ signaling is known to drive metastasis in human cancer. Under physiologic conditions, the level of TGFβ activity is tightly controlled by a regulatory network involving multiple negative regulators. At metastasis, however, these inhibitory mechanisms are usually overridden so that oncogenic TGFβ signaling can be overactivated and sustained. To better understand how the TGFβ inhibitors are suppressed in metastatic breast cancer cells, we compared miRNA expression profiles between breast cancers with or without metastasis and found that the miR424-503 cluster was markedly overexpressed in metastatic breast cancer. Mechanistic studies revealed that miR424 and miR503 simultaneously suppressed Smad7 and Smurf2, two key inhibitory factors of TGFβ signaling, leading to enhanced TGFβ signaling and metastatic capability of breast cancer cells. Moreover, antagonizing miR424-503 in breast cancer cells suppressed metastasis in vivo and increased overall host survival. Interestingly, our study also found that heterogeneous expression of the miR424-503 cluster contributed to the heterogeneity of TGFβ activity levels in, and metastatic potential of, breast cancer cell subsets. Overall, our findings demonstrate a novel mechanism, mediated by elevated expression of the miR424-503 cluster, underlying TGFβ activation and metastasis of human breast cancer.
Collapse
Affiliation(s)
- Yun Li
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China. Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Chinese Ministry of Education, Guangzhou, Guangdong, China
| | - Wei Li
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China. Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Chinese Ministry of Education, Guangzhou, Guangdong, China
| | - Zhe Ying
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China. Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Chinese Ministry of Education, Guangzhou, Guangdong, China
| | - Han Tian
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China. Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Chinese Ministry of Education, Guangzhou, Guangdong, China
| | - Xun Zhu
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China. Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Chinese Ministry of Education, Guangzhou, Guangdong, China
| | - Jun Li
- Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Chinese Ministry of Education, Guangzhou, Guangdong, China. Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Mengfeng Li
- Department of Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China. Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Chinese Ministry of Education, Guangzhou, Guangdong, China.
| |
Collapse
|
73
|
Abstract
Skeletal lesions are a frequent complication of breast and prostate cancer and a hallmark of multiple myeloma. Endocrine and paracrine factors modulate various aspects of bone metastases, including tumour proliferation, skeletal susceptibility to tumour homing, the microenvironment needed to support tumour persistence, and the initiation of a vicious cycle between tumour and bone-resident cells that further promotes tumour growth. Endocrine changes, such as oestrogen or vitamin D deficiency, contribute to a fertile bone microenvironment that might promote bone metastases. Bone health could be impaired further by existing cancer treatments, especially sex hormone deprivation. In this Review, we discuss the effect of hormones and associated local factors on cross-talk between bone metabolism and tumour biology. We review the biology of osteolytic and osteosclerotic lesions, with a focus on endocrine aspects, and outline potential therapeutic targets. We also summarise endocrine aspects of the pathogenesis and clinical presentation of bone metastases and provide an update on existing and future treatments.
Collapse
Affiliation(s)
- Lorenz C Hofbauer
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, TU Dresden, Dresden, Germany; Centre for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany.
| | - Tilman D Rachner
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, TU Dresden, Dresden, Germany
| | - Robert E Coleman
- Sheffield Cancer Research Centre, Weston Park Hospital, Sheffield, UK
| | - Franz Jakob
- Orthopaedic Center for Musculoskeletal Research, Wuerzburg, Germany
| |
Collapse
|
74
|
Guo F, Parker Kerrigan BC, Yang D, Hu L, Shmulevich I, Sood AK, Xue F, Zhang W. Post-transcriptional regulatory network of epithelial-to-mesenchymal and mesenchymal-to-epithelial transitions. J Hematol Oncol 2014; 7:19. [PMID: 24598126 PMCID: PMC3973872 DOI: 10.1186/1756-8722-7-19] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 02/27/2014] [Indexed: 12/26/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) and its reverse process, mesenchymal-to-epithelial transition (MET), play important roles in embryogenesis, stem cell biology, and cancer progression. EMT can be regulated by many signaling pathways and regulatory transcriptional networks. Furthermore, post-transcriptional regulatory networks regulate EMT; these networks include the long non-coding RNA (lncRNA) and microRNA (miRNA) families. Specifically, the miR-200 family, miR-101, miR-506, and several lncRNAs have been found to regulate EMT. Recent studies have illustrated that several lncRNAs are overexpressed in various cancers and that they can promote tumor metastasis by inducing EMT. MiRNA controls EMT by regulating EMT transcription factors or other EMT regulators, suggesting that lncRNAs and miRNA are novel therapeutic targets for the treatment of cancer. Further efforts have shown that non-coding-mediated EMT regulation is closely associated with epigenetic regulation through promoter methylation (e.g., miR-200 or miR-506) and protein regulation (e.g., SET8 via miR-502). The formation of gene fusions has also been found to promote EMT in prostate cancer. In this review, we discuss the post-transcriptional regulatory network that is involved in EMT and MET and how targeting EMT and MET may provide effective therapeutics for human disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fengxia Xue
- Department of Pathology, Unit 85, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA.
| | | |
Collapse
|
75
|
Kovanecz I, Gelfand R, Masouminia M, Gharib S, Segura D, Vernet D, Rajfer J, Li DK, Kannan K, Gonzalez-Cadavid NF. Oral Bisphenol A (BPA) given to rats at moderate doses is associated with erectile dysfunction, cavernosal lipofibrosis and alterations of global gene transcription. Int J Impot Res 2014; 26:67-75. [PMID: 24305612 PMCID: PMC4098849 DOI: 10.1038/ijir.2013.37] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 10/08/2013] [Accepted: 10/15/2013] [Indexed: 12/15/2022]
Abstract
Bisphenol A (BPA), a suspected reproductive biohazard and endocrine disruptor, released from plastics is associated with ED in occupationally exposed workers. However, in rats, despite the induction of hypogonadism, apoptosis of the penile corporal smooth muscle (SM), fat infiltration into the cavernosal tissue and changes in global gene expression with the intraperitoneal administration of high dose BPA, ED was not observed. We investigated whether BPA administered orally rather than intraperitoneally to rats for longer periods and lower doses will lead to ED. Main outcome measures are ED, histological, and biochemical markers in rat penile tissues. In all, 2.5-month-old rats were given drinking water daily without and with BPA at 1 and 0.1 mg kg(-1) per day. Two months later, erectile function was determined by cavernosometry and electrical field stimulation (EFS) and serum levels of testosterone (T), estradiol (E2) and BPA were measured. Penile tissue sections were assayed by Masson (SM/collagen), Oil Red O (fat), terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) (apoptosis), immunohistochemistry for Oct4 (stem cells), and α-SM actin/calponin (SM and myofibroblasts), applying quantitative image analysis. Other markers were assayed by western blotting. DNA microarrays/microRNA (miR) assays defined transcription profiles. Orally administered BPA did not affect body weight, but (1) decreased serum T and E2; (2) reduced the EFS response and increased the drop rate; (3) increased within the corporal tissue the presence of fat, myofibroblasts and apoptosis; (4) lowered the contents of SM and stem cells, but not nerve terminals; and (5) caused alterations in the transcriptional profiles for both mRNA and miRs within the penile shaft. Long-term exposure of rats to oral BPA caused a moderate corporal veno-occlusive dysfunction (CVOD), possibly due to alterations within the corporal tissue that pose gene transcriptional changes related to inflammation, fibrosis and epithelial/mesenchymal transition (EMT).
Collapse
Affiliation(s)
- I Kovanecz
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - R Gelfand
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA
- Division of Endocrinology, Charles Drew University of Medicine and Science, Los Angeles, CA
| | - M Masouminia
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA
| | - S Gharib
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA
| | - D Segura
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA
| | - D Vernet
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA
- Division of Endocrinology, Charles Drew University of Medicine and Science, Los Angeles, CA
| | - J Rajfer
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - DK Li
- Department of Health Research and Policy, Stanford University, Stanford, CA
- Division of Research, Kaiser Permanente
| | - K Kannan
- Wadsworth Center, New York State Department of Health, Albany, NY
| | - NF Gonzalez-Cadavid
- Division of Urology, Department of Surgery, Harbor-UCLA Medical Center and Los Angeles Biomedical Research Institute, Torrance, CA
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Division of Endocrinology, Charles Drew University of Medicine and Science, Los Angeles, CA
| |
Collapse
|
76
|
Jin L, Chen J, Li L, Li C, Chen C, Li S. CRH suppressed TGFβ1-induced Epithelial-Mesenchymal Transition via induction of E-cadherin in breast cancer cells. Cell Signal 2014; 26:757-65. [PMID: 24412750 DOI: 10.1016/j.cellsig.2013.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 12/29/2013] [Indexed: 01/22/2023]
Abstract
Since its discovery in biopsies from breast cancer patients, the effect of corticotropin-releasing hormone (CRH) on carcinoma progression is still unclear. Transforming growth factorβ1 (TGFβ1) promotes Epithelial-Mesenchymal Transition (EMT) and induces Snail1 and Twist1 expressions. Loss of epithelial cadherin (E-cadherin) mainly repressed by Snail1 and Twist1, has been considered as hallmark of Epithelial-Mesenchymal Transition (EMT). Two breast cancer cell lines, MCF-7 and MDA-MB-231 were used to investigate the effect of CRH on TGFβ1-induced EMT by transwell chamber. And HEK293 cells were transiently transfected with CRHR1 or CRHR2 to explore the definite effects of CRH receptor. We reported that CRH inhibited migration of human breast cancer cells through downregulation of Snail1 and Twist1, and subsequent upregulation of E-cadherin. CRH inhibited TGFβ1-mediated migration of MCF-7 via both CRHR1 and CRHR2 while this inhibition in MDA-MB-231 was mainly via CRHR2. Ectopic re-expression of CRHR1 or CRHR2 respectively in HEK293 cells increased E-cadherin expression after CRH stimulation. Furthermore, CRH repressed expression of mesenchymal marker, N-cadherin and induced expression of Occludin, inhibiting EMT in MCF-7 & MDA-MB-231. Our results suggest that CRH may function as a tumor suppressor, at least partly by regulating TGFβ1-mediated EMT. These results may contribute to uncovering the effect of CRH in breast tumorigenesis and progression.
Collapse
Affiliation(s)
- Lai Jin
- Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Department of Pharmacology, Nanjing Medical University, Nanjing 210029, China
| | - Jiandong Chen
- Department of Internal Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Li Li
- Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Department of Pharmacology, Nanjing Medical University, Nanjing 210029, China
| | - Chuanhua Li
- Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Department of Pharmacology, Nanjing Medical University, Nanjing 210029, China
| | - Cheng Chen
- Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Department of Pharmacology, Nanjing Medical University, Nanjing 210029, China
| | - Shengnan Li
- Jiangsu Provincial Key Lab of Cardiovascular Diseases and Molecular Intervention, Department of Pharmacology, Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
77
|
Balogh P, Szabó A, Katz S, Likó I, Patócs A, L.Kiss A. Estrogen receptor alpha is expressed in mesenteric mesothelial cells and is internalized in caveolae upon Freund's adjuvant treatment. PLoS One 2013; 8:e79508. [PMID: 24244516 PMCID: PMC3828353 DOI: 10.1371/journal.pone.0079508] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 09/27/2013] [Indexed: 12/03/2022] Open
Abstract
Transformation of epithelial cells into connective tissue cells (epithelial-mesenchymal transition, EMT) is a complex mechanism involved in tumor metastasis, and in normal embryogenesis, while type II EMT is mainly associated with inflammatory events and tissue regenaration. In this study we examined type II EMT at the ultrastructural and molecular level during the inflammatory process induced by Freund's adjuvant treatment in rat mesenteric mesothelial cells. We found that upon the inflammatory stimulus mesothelial cells lost contact with the basal lamina and with each other, and were transformed into spindle-shaped cells. These morphological changes were accompanied by release of interleukins IL-1alpha, -1beta and IL-6 and by secretion of transforming growth factor beta (TGF-β) into the peritoneal cavity. Mesothelial cells also expressed estrogen receptor alpha (ER-α) as shown by immunolabeling at the light and electron microscopical levels, as well as by quantitative RT-PCR. The mRNA level of ER-α showed an inverse correlation with the secretion of TGF-β. At the cellular and subcellular levels ER-α was colocalized with the coat protein caveolin-1 and was found in the plasma membrane of mesothelial cells, in caveolae close to multivesicular bodies (MVBs) or in the membrane of these organelles, suggesting that ER-α is internalized via caveola-mediated endocytosis during inflammation. We found asymmetric, thickened, electron dense areas on the limiting membrane of MVBs (MVB plaques) indicating that these sites may serve as platforms for collecting and organizing regulatory proteins. Our morphological observations and biochemical data can contribute to form a potential model whereby ER-α and its caveola-mediated endocytosis might play role in TGF-β induced type II EMT in vivo.
Collapse
Affiliation(s)
- Petra Balogh
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, Hungary
- * E-mail:
| | - Arnold Szabó
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Sándor Katz
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - István Likó
- Pharmacology and Drug Safety Research, R. Gedeon Plc, Hungary
| | - Attila Patócs
- HSA-SE Lendület Hereditary Endocrine Tumors Research Group, Budapest, Hungary
| | - Anna L.Kiss
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
78
|
Zhang X, Zhang B, Gao J, Wang X, Liu Z. Regulation of the microRNA 200b (miRNA-200b) by transcriptional regulators PEA3 and ELK-1 protein affects expression of Pin1 protein to control anoikis. J Biol Chem 2013; 288:32742-32752. [PMID: 24072701 DOI: 10.1074/jbc.m113.478016] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
MicroRNA (miRNA) 200s regulate E-cadherin by directly targeting ZEB1/ZEB2, which are transcriptional repressors of E-cadherin. Decreased expression of E-cadherin results in cancer cells losing interaction with the extracellular matrix and detaching from the primary tumor. Normally, cells will undergo anoikis after losing interaction with the extracellular matrix. Cancer cells must, therefore, possess the ability to resist anoikis during the process of metastasis. Here we show that miRNA-200b regulates anoikis by directly targeting the 3' UTR of Pin1 mRNA and regulating Pin1 expression at the translational level. We found that down-regulation of miRNA-200b promotes cancer cells survival during metastasis, and the homeless state of these cells resulted in decreased expression of miRNA-200b in the MCF-7 cell line. We also found that expression of miRNA-200b is down-regulated in human breast cancer during lymph node metastasis, which has a significant negative correlation with Pin1 expression. Two members of the ETS (E-26) family (PEA3 and ELK-1) regulate the expression of miRNA-200b. PEA3 promotes the expression of miRNA-200b, and ELK-1 is a transcriptional repressor of miRNA-200b. In addition, miRNA-200b regulates the activity of PEA3 and ELK-1 via the Pin1-pERK pathway and forms self-regulated feedback loops. This study characterizes the role of miRNA-200b in the regulation of anoikis and demonstrates the regulation of its own expression in the process of metastasis.
Collapse
Affiliation(s)
- Xusen Zhang
- From the State Key Laboratory of Molecular Oncology
| | - Bailin Zhang
- Department of Abdominal Surgery, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jidong Gao
- Department of Abdominal Surgery, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiang Wang
- Department of Abdominal Surgery, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhihua Liu
- From the State Key Laboratory of Molecular Oncology.
| |
Collapse
|
79
|
Cvetković D, Babwah AV, Bhattacharya M. Kisspeptin/KISS1R System in Breast Cancer. J Cancer 2013; 4:653-61. [PMID: 24155777 PMCID: PMC3805993 DOI: 10.7150/jca.7626] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 09/22/2013] [Indexed: 01/18/2023] Open
Abstract
Kisspeptins (KP), peptide products of the kisspeptin-1 (KISS1) gene are the endogenous ligands for a G protein-coupled receptor (GPCR) - KP receptor (KISS1R). KISS1R couples to the Gαq/11 signaling pathway. KISS1 is a metastasis suppressor gene and the KP/KISS1R signaling has anti-metastatic and tumor-suppressant effects in numerous human cancers. On the other hand, recent studies indicate that KP/KISS1R pathway plays detrimental roles in breast cancer. In this review, we summarize recent developments in the understanding of the mechanisms regulating KP/KISS1R signaling in breast cancer metastasis.
Collapse
Affiliation(s)
- Donna Cvetković
- 1. Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada, N6A 5C1
| | | | | |
Collapse
|
80
|
Epigenetic regulation of hepatocellular carcinoma in non-alcoholic fatty liver disease. Semin Cancer Biol 2013; 23:471-82. [PMID: 24018165 DOI: 10.1016/j.semcancer.2013.08.010] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 08/30/2013] [Indexed: 02/07/2023]
Abstract
Emerging evidence that epigenetics converts alterations in nutrient and metabolism into heritable pattern of gene expression has profound implications in understanding human physiology and diseases. Non-alcoholic fatty liver disease (NAFLD) is the hepatic manifestation of metabolic syndrome including obesity and diabetes which elevate the risk of hepatocellular carcinoma (HCC) especially in male. This review focuses on the molecular connections between metabolic dysfunction and aberrant epigenetic alterations in the development of HCC in NAFLD. The metabolites derived from excessive insulin, glucose and lipid may perturb epigenetic gene regulation through DNA methylation, histone modifications, and RNA interference, leading to activation of pro-inflammatory signaling and deregulation of metabolic pathways. The interplay and crosstalk of chromatin-modifying enzymes, microRNAs, signaling pathways and the downstream transcription factors result in epigenomic reprogramming that drives hepatocellular transformation. The interactions between sex hormone pathways and the epigenetic machineries that influence chromatin states in NAFLD provide potential molecular mechanisms of gender disparity in HCC. A deeper understanding of these connections and comprehensive molecular catalog of hepatocarcinogenesis may shed light in the identification of druggable epigenetic targets for the prevention and treatment of HCC in obese or diabetic patients.
Collapse
|
81
|
Cai ZG, Zhang SM, Zhang H, Zhou YY, Wu HB, Xu XP. Aberrant expression of microRNAs involved in epithelial-mesenchymal transition of HT-29 cell line. Cell Biol Int 2013; 37:669-74. [PMID: 23483606 DOI: 10.1002/cbin.10087] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Accepted: 02/08/2013] [Indexed: 02/02/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is an essential step for cancer metastasis. MicroRNAs (miRNAs) are small non-coding RNAs that regulate target-mRNAs post-transcriptionally. The expression and function of miRNAs in EMT of HT-29 colonic cells remain elusive. This study looks at expression of miRNAs in EMT and explores the effects of miRNAs on EMT in HT-29 cell line. HT-29 was treated with TGF β to establish an EMT model, in which a collection of miRNAs was dynamically regulated by real-time PCR (qPCR) analysis. Among them, miR-21 and miR-27 were significantly upregulated, while miR-22, miR-26, miR-30, miR-181, miR-200b, miR-200c and miR-214 were markedly downregulated. MiRNA-inhibitors were used to knockdown miRNAs in HT-29 and EMT markers were determined by qPCR to monitor the effects of miRNAs on EMT process. Results showed that miR-22 could not alter the expression of EMT markers, while knockdown of miR-200b could significantly increase that of epithelial markers, N-cadherin, Vimentin, α-Sma and Twist1 and decrease that of mesenchymal marker, E-cadherin. Bioinformatic analysis and Western blot showed that ZEB1 was directly suppressed by miR-200b. In conclusion, miRNAs are dynamically regulated in TGF β-induced EMT of HT-29 and miR-200b was essential for EMT by suppressing the expression of ZEB1 in HT-29.
Collapse
Affiliation(s)
- Zhi-Gang Cai
- Department of Cardio-Thoracic Surgery, Number 455 Hospital of The Chinese People's Liberation Army, Shanghai 200052, China.
| | | | | | | | | | | |
Collapse
|
82
|
He GH, Lu J, Shi PP, Xia W, Yin SJ, Jin TB, Chen DD, Xu GL. Polymorphisms of human histamine receptor H4 gene are associated with breast cancer in Chinese Han population. Gene 2013; 519:260-265. [PMID: 23481304 DOI: 10.1016/j.gene.2013.02.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Accepted: 02/11/2013] [Indexed: 12/21/2022]
Abstract
Previous investigations indicated that histamine receptor H4 (HRH4) played important roles in many aspects of breast cancer pathogenesis, and that the polymorphisms of HRH4 gene may result in expression and functional changes of HRH4 proteins. However, the relationship between polymorphisms of HRH4 and breast cancer risk and malignant degree is unclear. In the present study, we conducted a case-control investigation among 185 Chinese Han breast cancer patients and 199 ethnicity-matched health controls. Four tag-SNPs (i.e. rs623590, rs16940762, rs11662595 and rs1421125) of HRH4 were genotyped and association analysis was performed. Odds ratios (ORs) with 95% confidence intervals (CI) were used to assess the association. We found that the T allele of rs623590 had a decreased risk of breast cancer (adjusted OR, 0.667; 95% CI, 0.486-0.913; P=0.012) while the A allele of rs1421125 had an increased risk (adjusted OR, 1.653; 95% CI, 1.139-2.397; P=0.008). Further haplotype analysis showed that the CAA haplotype of rs623590-rs11662595-rs1421125 was more frequent among patients with breast cancer (adjusted OR, 1.856; 95% CI, 1.236-2.787; P=0.003). Additionally, polymorphisms of rs623590 and rs11662595 were also correlated with clinical stages, lymph node involvement, and HER2 status. These findings indicated that the variants of rs623590, rs11662595 and rs1421125 genotypes of HRH4 gene were significantly associated with the risk and malignant degree of breast cancer in Chinese Han populations, which may provide us novel insight into the pathogenesis of breast cancer although further studies with larger participants worldwide are still needed for conclusion validation.
Collapse
Affiliation(s)
- Gong-Hao He
- Department of Pharmacy, Kunming General Hospital of Chengdu Military Region, Kunming 650032, China
| | | | | | | | | | | | | | | |
Collapse
|
83
|
Manavalan TT, Teng Y, Litchfield LM, Muluhngwi P, Al-Rayyan N, Klinge CM. Reduced expression of miR-200 family members contributes to antiestrogen resistance in LY2 human breast cancer cells. PLoS One 2013; 8:e62334. [PMID: 23626803 PMCID: PMC3633860 DOI: 10.1371/journal.pone.0062334] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 03/20/2013] [Indexed: 02/02/2023] Open
Abstract
INTRODUCTION The role of miRNAs in acquired endocrine-resistant breast cancer is not fully understood. One hallmark of tumor progression is epithelial-to-mesenchymal transition (EMT), characterized by a loss of cell adhesion resulting from reduced E-cadherin and increased cell mobility. miR-200 family members regulate EMT by suppressing expression of transcriptional repressors ZEB1/2. Previously we reported that the expression of miR-200a, miR-200b, and miR-200c was lower in LY2 endocrine-resistant, mesenchymal breast cancer cells compared to parental, endocrine sensitive, epithelial MCF-7 breast cancer cells. Here we investigated the regulation of miR-200 family members and their role in endocrine-sensitivity in breast cancer cells. RESULTS miR-200 family expression was progressively reduced in a breast cancer cell line model of advancing endocrine/tamoxifen (TAM) resistance. Concomitant with miR-200 decrease, there was an increase in ZEB1 mRNA expression. Overexpression of miR-200b or miR-200c in LY2 cells altered cell morphology to a more epithelial appearance and inhibited cell migration. Further, miR-200b and miR-200c overexpression sensitized LY2 cells to growth inhibition by estrogen receptor (ER) antagonists TAM and fulvestrant. Knockdown of ZEB1 in LY2 cells recapitulated the effect of miR-200b and miR-200c overexpression resulting in inhibition of LY2 cell proliferation by TAM and fulvestrant, but not the aromatase inhibitor exemestane. Demethylating agent 5-aza-2'-deoxycytidine (5-aza-dC) in combination with histone deacetylase inhibitor trichostatin A (TSA) increased miR-200b and miR-200c in LY2 cells. Concomitant with the increase in miR-200b and miR-200c, ZEB1 expression was decreased and cells appeared more epithelial in morphology and were sensitized to TAM and fulvestrant inhibition. Likewise, knockdown of ZEB1 increased antiestrogen sensitivity of LY2 cells resulting in inhibition of cell proliferation. CONCLUSIONS Our data indicate that reduced miRNA-200b and miR-200c expression contributes to endocrine resistance in breast cancer cells and that the reduced expression of these miR-200 family members in endocrine-resistant cells can be reversed by 5-aza-dC+TSA.
Collapse
Affiliation(s)
- Tissa T. Manavalan
- Department of Biochemistry and Molecular Biology, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Yun Teng
- Department of Biochemistry and Molecular Biology, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Lacey M. Litchfield
- Department of Biochemistry and Molecular Biology, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Penn Muluhngwi
- Department of Biochemistry and Molecular Biology, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Numan Al-Rayyan
- Department of Biochemistry and Molecular Biology, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Carolyn M. Klinge
- Department of Biochemistry and Molecular Biology, Center for Genetics and Molecular Medicine, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| |
Collapse
|
84
|
Zhang X, Li Y, Zhang Y, Song J, Wang Q, Zheng L, Liu D. Beta-elemene blocks epithelial-mesenchymal transition in human breast cancer cell line MCF-7 through Smad3-mediated down-regulation of nuclear transcription factors. PLoS One 2013; 8:e58719. [PMID: 23516540 PMCID: PMC3597725 DOI: 10.1371/journal.pone.0058719] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 02/05/2013] [Indexed: 01/23/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is the first step required for breast cancer to initiate metastasis. However, the potential of drugs to block and reverse the EMT process are not well explored. In the present study, we investigated the inhibitory effect of beta-elemene (ELE), an active component of a natural plant-derived anti-neoplastic agent in an established EMT model mediated by transforming growth factor-beta1 (TGF-β1). We found that ELE (40 µg/ml ) blocked the TGF-β1-induced phenotypic transition in the human breast cancer cell line MCF-7. ELE was able to inhibit TGF-β1-mediated upregulation of mRNA and protein expression of nuclear transcription factors (SNAI1, SNAI2, TWIST and SIP1), potentially through decreasing the expression and phosphorylation of Smad3, a central protein mediating the TGF-β1 signalling pathway. These findings suggest a potential therapeutic benefit of ELE in treating basal-like breast cancer.
Collapse
Affiliation(s)
- Xian Zhang
- Department of Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
- Research Institute of Integrated Traditional and Western Medicine of Dalian Medical University, Dalian, China
| | - Yinghua Li
- Department of Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yang Zhang
- Department of Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
- * E-mail:
| | - Jincheng Song
- Graduate Institute of Dalian Medical University, Dalian, China
| | - Qimin Wang
- Department of Pathology, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Luping Zheng
- Research Institute of Integrated Traditional and Western Medicine of Dalian Medical University, Dalian, China
| | - Dan Liu
- Graduate Institute of Dalian Medical University, Dalian, China
| |
Collapse
|
85
|
Cong N, Du P, Zhang A, Shen F, Su J, Pu P, Wang T, Zjang J, Kang C, Zhang Q. Downregulated microRNA-200a promotes EMT and tumor growth through the wnt/β-catenin pathway by targeting the E-cadherin repressors ZEB1/ZEB2 in gastric adenocarcinoma. Oncol Rep 2013; 29:1579-87. [PMID: 23381389 DOI: 10.3892/or.2013.2267] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 12/28/2012] [Indexed: 01/12/2023] Open
Abstract
In a previous study, we found that microRNA (miRNA)-200a suppresses Wnt/β-catenin signaling by interacting with β-catenin, thereby inhibiting migration, invasion and proliferation. However, the mechanism involved in this suppression remains unclear. In the present study, we investigated the underlying mechanism of miR-200a regulation of epithelial-mesenchymal transition (EMT) in gastric carcinoma cells, and confirmed the tumor suppressor role of miR-200a in vivo. The expressions of miRNA-200a, -200b and -200c, identified by fluorescent in situ hybridization, were downregulated and inversely correlated with WHO grades of gastric adenocarcinoma (GA). The expression of the potential miR-200a target genes ZEB1 and ZEB2 was detected immunohistochemically. These examinations used the same tissue microarrays to analyze the relationships between miR-200a and potential target genes. The expression of miR-200a and ZEB1/ZEB2 in the same GA tissue microarrays was inversely related. Restored miR-200a expression inhibited tumor growth in nude mice harboring subcutaneous SGC7901 xenografts. The expression of N-cadherin, β-catenin, Twist1 and Snail2 decreased, and E-cadherin levels increased, when miR-200a was elevated, as tested by fluorescence microscopy and immunohistochemistry. Similar results were observed in vivo. We found upregulated miR-200a expression to increase E-cadherin and suppress the Wnt/β-catenin pathway by targeting ZEB1 and ZEB2 in GA, thus delaying tumor growth in vivo. The effect of miR-200a on Wnt/β-catenin signaling may provide a therapeutic target against EMT.
Collapse
Affiliation(s)
- Ningning Cong
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Waldmeier L, Meyer-Schaller N, Diepenbruck M, Christofori G. Py2T murine breast cancer cells, a versatile model of TGFβ-induced EMT in vitro and in vivo. PLoS One 2012; 7:e48651. [PMID: 23144919 PMCID: PMC3492491 DOI: 10.1371/journal.pone.0048651] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 09/26/2012] [Indexed: 01/05/2023] Open
Abstract
Introduction Increasing evidence supports a role of an epithelial to mesenchymal transition (EMT) process in endowing subsets of tumor cells with properties driving malignant tumor progression and resistance to cancer therapy. To advance our understanding of the underlying mechanisms, we sought to generate a transplantable cellular model system that allows defined experimental manipulation and analysis of EMT in vitro and at the same time recapitulates oncogenic EMT in vivo. Methodology/Results We have established a stable murine breast cancer cell line (Py2T) from a breast tumor of an MMTV-PyMT transgenic mouse. Py2T cells display a metastable epithelial phenotype characterized by concomitant expression of luminal and basal cytokeratins and sheet migration. Exposure of Py2T cells to transforming growth factor β (TGFβ) in vitro induces reversible EMT accompanied by downregulation of E-cadherin and upregulation of mesenchymal markers, including EMT transcription factors, and a gain in single cell motility and invasiveness. Py2T cells give rise to tumors after orthotopic injection into syngeneic FVB/N mice. Notably, transplantation of epithelial Py2T cells results in the formation of invasive primary tumors with low to absent E-cadherin expression, indicating that the cells undergo EMT-like changes in vivo. This process appears to at least in part depend on TGFβ signaling, since tumors formed by Py2T cells expressing a dominant-negative version of TGFβ receptor widely maintain their epithelial differentiation status. Conclusions/Significance Together, the data demonstrate that the Py2T cell line represents a versatile model system to study the EMT process in vitro and in vivo. The observation that Py2T cells give rise to tumors and collectively undergo EMT-like changes in vivo highlights the suitability of the Py2T model system as a tool to study tumor-related EMT. In particular, Py2T cells may serve to corroborate recent findings relating EMT to cancer cell stemness, to therapy resistance and to tumor recurrence.
Collapse
Affiliation(s)
- Lorenz Waldmeier
- Institute of Biochemistry and Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Nathalie Meyer-Schaller
- Institute of Biochemistry and Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Maren Diepenbruck
- Institute of Biochemistry and Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Gerhard Christofori
- Institute of Biochemistry and Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland
- * E-mail:
| |
Collapse
|
87
|
Abstract
Estrogen exhibits a broad spectrum of physiological functions ranging from regulation of the menstrual cycle and reproduction to modulation of bone density, brain function, and cholesterol mobilization. Despite the beneficial actions of endogenous estrogen, sustained exposure to exogenous estrogen is a well-established risk factor for various cancers. We summarize our current understanding of the molecular mechanisms of estrogen signaling in normal and cancer cells and discuss the major challenges to existing antiestrogen therapies.
Collapse
Affiliation(s)
- Jing Liang
- Tianjin Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin 300070, China.
| | | |
Collapse
|
88
|
Lambertini E, Lolli A, Vezzali F, Penolazzi L, Gambari R, Piva R. Correlation between Slug transcription factor and miR-221 in MDA-MB-231 breast cancer cells. BMC Cancer 2012; 12:445. [PMID: 23031797 PMCID: PMC3534407 DOI: 10.1186/1471-2407-12-445] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 09/28/2012] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Breast cancer and its metastatic progression is mainly directed by epithelial to mesenchymal transition (EMT), a phenomenon supported by specific transcription factors and miRNAs. METHODS In order to investigate a possible correlation between Slug transcription factor and miR-221, we performed Slug gene silencing in MDA-MB-231 breast cancer cells and evaluated the expression of genes involved in supporting the breast cancer phenotype, using qRT-PCR and Western blot analysis. Chromatin immunoprecipitation and wound healing assays were employed to determine a functional link between these two molecules. RESULTS We showed that Slug silencing significantly decreased the level of miR-221 and vimentin, reactivated Estrogen Receptor α and increased E-cadherin and TRPS1 expression. We demonstrated that miR-221 is a Slug target gene, and identified a specific region of miR-221 promoter that is transcriptionally active and binds the transcription factor Slug "in vivo". In addition, we showed that in Slug-silenced cells, wich retained residual miR-221 (about 38%), cell migration was strongly inhibited. Cell migration was inhibited, but to a less degree, following complete knockdown of miR-221 expression by transfection with antagomiR-221. CONCLUSIONS We report for the first time evidence of a correlation between Slug transcription factor and miR-221 in breast cancer cells. These studies suggest that miR-221 expression is, in part, dependent on Slug in breast cancer cells, and that Slug plays a more important role than miR-221 in cell migration and invasion.
Collapse
Affiliation(s)
- Elisabetta Lambertini
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, 44121, Italy
| | | | | | | | | | | |
Collapse
|
89
|
Kiesslich T, Pichler M, Neureiter D. Epigenetic control of epithelial-mesenchymal-transition in human cancer. Mol Clin Oncol 2012; 1:3-11. [PMID: 24649114 DOI: 10.3892/mco.2012.28] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2012] [Accepted: 09/07/2012] [Indexed: 02/06/2023] Open
Abstract
Development and tissue homeostasis as well as carcinogenesis share the evolutionary conserved process of epithelial-mesenchymal transition (EMT). EMT enables differentiated epithelial cells to trans-differentiate to a mesenchymal phenotype which is associated with diverse cellular properties including altered morphology, migration and invasion and stemness. In physiological development and tissue homeostasis, EMT exerts beneficial functions for structured tissue formation and maintenance. Under pathological conditions, EMT causes uncontrolled tissue repair and organ fibrosis, as well as the induction of tumor growth, angiogenesis and metastasis in the context of cancer progression. Particularly, the metastatic process is essentially linked to diverse EMT-driven functions which give the mesenchymal differentiated tumor cells the capacity to migrate and form micrometastases in distant organs. Recent analyses of the mechanisms controlling EMT revealed a significant epigenetic regulatory impact reflecting the reversible nature of EMTs. As several approaches of epigenetic therapy are already under clinical evaluation, including inhibitors of DNA methyl transferase and histone deacetylase, targeting the epigenetic regulation of EMT may represent a promising therapeutic option in the future. Therefore, we undertook this review to reassess the current knowledge on the roles of epigenetic control in the regulation of EMT in human cancer. These recent findings are discussed in view of their implications on future diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Tobias Kiesslich
- Institute of Pathology; ; Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), Salzburg
| | - Martin Pichler
- Division of Oncology, Department of Internal Medicine, Medical University of Graz (MUG), Graz, Austria
| | | |
Collapse
|
90
|
Sokolosky ML, Wargovich MJ. Homeostatic imbalance and colon cancer: the dynamic epigenetic interplay of inflammation, environmental toxins, and chemopreventive plant compounds. Front Oncol 2012; 2:57. [PMID: 22675672 PMCID: PMC3365481 DOI: 10.3389/fonc.2012.00057] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 05/14/2012] [Indexed: 12/18/2022] Open
Abstract
The advent of modern medicine has allowed for significant advances within the fields of emergency care, surgery, and infectious disease control. Health threats that were historically responsible for immeasurable tolls on human life are now all but eradicated within certain populations, specifically those that enjoy higher degrees of socio-economic status and access to healthcare. However, modernization and its resulting lifestyle trends have ushered in a new era of chronic illness; one in which an unprecedented number of people are estimated to contract cancer and other inflammatory diseases. Here, we explore the idea that homeostasis has been redefined within just a few generations, and that diseases such as colorectal cancer are the result of fluctuating physiological and molecular imbalances. Phytochemical-deprived, pro-inflammatory diets combined with low-dose exposures to environmental toxins, including bisphenol-A (BPA) and other endocrine disruptors, are now linked to increasing incidences of cancer in westernized societies and developing countries. There is recent evidence that disease determinants are likely set in utero and further perpetuated into adulthood dependent upon the innate and environmentally-acquired phenotype unique to each individual. In order to address a disease as multi-factorial, case-specific, and remarkably adaptive as cancer, research must focus on its root causes in order to elucidate the molecular mechanisms by which they can be prevented or counteracted via plant-derived compounds such as epigallocatechin-3-gallate (EGCG) and resveratrol. The significant role of epigenetics in the regulation of these complex processes is emphasized here to form a comprehensive view of the dynamic interactions that influence modern-day carcinogenesis, and how sensibly restoring homeostatic balance may be the key to the cancer riddle.
Collapse
Affiliation(s)
- Melissa L Sokolosky
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina Charleston, SC, USA
| | | |
Collapse
|