151
|
Modulation of Estrogen Response Element-Driven Gene Expressions and Cellular Proliferation with Polar Directions by Designer Transcription Regulators. PLoS One 2015; 10:e0136423. [PMID: 26295471 PMCID: PMC4546503 DOI: 10.1371/journal.pone.0136423] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 08/03/2015] [Indexed: 12/27/2022] Open
Abstract
Estrogen receptor α (ERα), as a ligand-dependent transcription factor, mediates 17β-estradiol (E2) effects. ERα is a modular protein containing a DNA binding domain (DBD) and transcription activation domains (AD) located at the amino- and carboxyl-termini. The interaction of the E2-activated ERα dimer with estrogen response elements (EREs) of genes constitutes the initial step in the ERE-dependent signaling pathway necessary for alterations of cellular features. We previously constructed monomeric transcription activators, or monotransactivators, assembled from an engineered ERE-binding module (EBM) using the ERα-DBD and constitutively active ADs from other transcription factors. Monotransactivators modulated cell proliferation by activating and repressing ERE-driven gene expressions that simulate responses observed with E2-ERα. We reasoned here that integration of potent heterologous repression domains (RDs) into EBM could generate monotransrepressors that alter ERE-bearing gene expressions and cellular proliferation in directions opposite to those observed with E2-ERα or monotransactivators. Consistent with this, monotransrepressors suppressed reporter gene expressions that emulate the ERE-dependent signaling pathway. Moreover, a model monotransrepressor regulated DNA synthesis, cell cycle progression and proliferation of recombinant adenovirus infected ER-negative cells through decreasing as well as increasing gene expressions with polar directions compared with E2-ERα or monotransactivator. Our results indicate that an ‘activator’ or a ‘repressor’ possesses both transcription activating/enhancing and repressing/decreasing abilities within a chromatin context. Offering a protein engineering platform to alter signal pathway-specific gene expressions and cell growth, our approach could also be used for the development of tools for epigenetic modifications and for clinical interventions wherein multigenic de-regulations are an issue.
Collapse
|
152
|
Kim SH, Madak-Erdogan Z, Bae SC, Carlson KE, Mayne CG, Granick S, Katzenellenbogen BS, Katzenellenbogen JA. Ligand Accessibility and Bioactivity of a Hormone-Dendrimer Conjugate Depend on pH and pH History. J Am Chem Soc 2015; 137:10326-35. [PMID: 26186415 DOI: 10.1021/jacs.5b05952] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Estrogen conjugates with a polyamidoamine (PAMAM) dendrimer have shown remarkably selective regulation of the nongenomic actions of estrogens in target cells. In response to pH changes, however, these estrogen-dendrimer conjugates (EDCs) display a major morphological transition that alters the accessibility of the estrogen ligands that compromises the bioactivity of the EDC. A sharp break in dynamic behavior near pH 7 occurs for three different ligands on the surface of a PAMAM-G6 dendrimer: a fluorophore (tetramethylrhodamine [TMR]) and two estrogens (17α-ethynylestradiol and diphenolic acid). Collisional quenching and time-resolved fluorescence anisotropy experiments with TMR-PAMAM revealed high ligand shielding above pH 7 and low shielding below pH 7. Furthermore, when the pH was cycled from 8.5 (conditions of ligand-PAMAM conjugation) to 4.5 (e.g., endosome/lysosome) and through 6.5 (e.g., hypoxic environment) back to pH 8.5, the 17α-ethynylestradiol- and diphenolic acid-PAMAM conjugates experienced a dramatic, irreversible loss in cell stimulatory activity; dynamic NMR studies indicated that the hormonal ligands had become occluded within the more hydrophobic core of the PAMAM dendrimer. Thus, the active state of these estrogen-dendrimer conjugates appears to be metastable. This pH-dependent irreversible masking of activity is of considerable relevance to the design of drug conjugates with amine-bearing PAMAM dendrimers.
Collapse
Affiliation(s)
| | | | - Sung Chul Bae
- §IBS Center for Soft and Living Matter and UNIST, Ulsan 689-798, South Korea
| | | | | | - Steve Granick
- §IBS Center for Soft and Living Matter and UNIST, Ulsan 689-798, South Korea
| | | | | |
Collapse
|
153
|
Perillo B, Di Santi A, Cernera G, Ombra MN, Castoria G, Migliaccio A. Nuclear receptor-induced transcription is driven by spatially and timely restricted waves of ROS. The role of Akt, IKKα, and DNA damage repair enzymes. Nucleus 2015; 5:482-91. [PMID: 25482200 PMCID: PMC4164490 DOI: 10.4161/nucl.36274] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gene expression is governed by chromatin mainly through posttranslational modifications at the N-terminal tails of nucleosomal histone proteins. According to the histone code theory, peculiar sets of such modifications (marks) give rise to reproducible final effects on transcription and, very recently, a further level of complexity has been highlighted in binary switches between specific marks at adjacent residues. In particular, disappearance of dimethyl-lysine 9 in histone H3 is faced by phosphorylation of the following serine during activation of gene expression. Demethylation of lysine 9 by the lysine-specific demethylase 1 (LSD1) is a pre-requisite for addition of the phosphoryl mark to serine 10 and an essential step in the transcriptional control by estrogens. It generates a local burst of oxygen reactive species (ROS) that induce oxidation of nearby nucleotides and recruitment of repair enzymes with a consequent formation of single or double stranded nicks on DNA that modify chromatin flexibility in order to allow correct assembly of the transcriptional machinery.
We describe here the molecular mechanism by which members of the family of nuclear receptors prevent the potential damage to DNA during transcription of target genes elicited by the use of ROS to shape chromatin. The mechanism is based on the presence of phosphorylated serine 10 in histone H3 to prevent unbalanced DNA oxidation waves. We also discuss the opportunities raised by the use of voluntary derangement of this servo system to induce selective death in hormone-responsive transformed cells.
Collapse
Affiliation(s)
- Bruno Perillo
- a Istituto di Scienze dell'Alimentazione; Avellino, Italy
| | | | | | | | | | | |
Collapse
|
154
|
Sánchez-Aguilera A, Arranz L, Martín-Pérez D, García-García A, Stavropoulou V, Kubovcakova L, Isern J, Martín-Salamanca S, Langa X, Skoda RC, Schwaller J, Méndez-Ferrer S. Estrogen signaling selectively induces apoptosis of hematopoietic progenitors and myeloid neoplasms without harming steady-state hematopoiesis. Cell Stem Cell 2015; 15:791-804. [PMID: 25479752 DOI: 10.1016/j.stem.2014.11.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 10/08/2014] [Accepted: 11/05/2014] [Indexed: 12/31/2022]
Abstract
Estrogens are potent regulators of mature hematopoietic cells; however, their effects on primitive and malignant hematopoietic cells remain unclear. Using genetic and pharmacological approaches, we observed differential expression and function of estrogen receptors (ERs) in hematopoietic stem cell (HSC) and progenitor subsets. ERα activation with the selective ER modulator (SERM) tamoxifen induced apoptosis in short-term HSCs and multipotent progenitors. In contrast, tamoxifen induced proliferation of quiescent long-term HSCs, altered the expression of self-renewal genes, and compromised hematopoietic reconstitution after myelotoxic stress, which was reversible. In mice, tamoxifen treatment blocked development of JAK2(V617F)-induced myeloproliferative neoplasm in vivo, induced apoptosis of human JAK2(V617F+) HSPCs in a xenograft model, and sensitized MLL-AF9(+) leukemias to chemotherapy. Apoptosis was selectively observed in mutant cells, and tamoxifen treatment only had a minor impact on steady-state hematopoiesis in disease-free animals. Together, these results uncover specific regulation of hematopoietic progenitors by estrogens and potential antileukemic properties of SERMs.
Collapse
Affiliation(s)
- Abel Sánchez-Aguilera
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain.
| | - Lorena Arranz
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Daniel Martín-Pérez
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Andrés García-García
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Vaia Stavropoulou
- Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Lucia Kubovcakova
- Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Joan Isern
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Sandra Martín-Salamanca
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Xavier Langa
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Radek C Skoda
- Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Jürg Schwaller
- Department of Biomedicine, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Simón Méndez-Ferrer
- Stem Cell Niche Pathophysiology Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain.
| |
Collapse
|
155
|
Affiliation(s)
- Olugbenga T. Awolaran
- Faculty of Health and Life Sciences, Oxford Brookes University, Jack Straws Lane, Marston, Oxford, OX3 0FL, UK
| |
Collapse
|
156
|
Herrmann P, Aronica SM. Estrogen and tamoxifen up-regulate FXYD3 on breast cancer cells: assessing the differential roles of ER α and ZEB1. SPRINGERPLUS 2015; 4:245. [PMID: 26090296 PMCID: PMC4467593 DOI: 10.1186/s40064-015-1022-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 05/11/2015] [Indexed: 11/27/2022]
Abstract
Increased expression of the FXYD3 family of proteins has been associated with lung, colorectal, bladder and pancreatic cancers, and recent evidence suggests that elevated FXYD3 may promote tumor cell proliferation in breast cancer as well. However, factors involved in up-regulating the expression of FXYD3 in breast cancer have not been identified. We evaluated whether estrogen and the selective estrogen receptor modulator tamoxifen could regulate the expression of FXYD3 on breast cancer cells. Estrogen receptor (ER) α-positive MCF-7 and ER α-negative MDA-MB-231 human breast cancer cells used in our studies were treated with estrogen, tamoxifen or the combination of these agents. Relative expression of FXYD3 was assessed using fluorochrome-tagged antibodies and a fluorescence cytometer. We found that estrogen and tamoxifen, used alone or in combination, significantly increased FXYD3 on MCF-7 cells. FXYD3 levels did not increase compared to the control samples when ER α-negative 231 cells were treated with estrogen or tamoxifen, alone or in combination, indicating that ER α was required for the increased FXYD3 response. We showed that ER α associates with the transcription factor ZEB1 in MCF-7 cells, and that decreasing ZEB1 protein expression using siRNA disrupts the ability of estrogen, but not tamoxifen, to increase FXYD3 in MCF-7 cells. Our results indicate that there may be two mechanisms, both involving ER α and one requiring ZEB1, through which FXYD3 may be increased by estrogen and tamoxifen in breast cancer cells. Ongoing research endeavors are focusing on identifying cellular components through which estrogen and tamoxifen, alone or in combination, differentially regulate FXYD3 expression in human breast cancer cells.
Collapse
Affiliation(s)
- Paul Herrmann
- Department of Biology, Canisius College, 2001 Main Street, Buffalo, NY 14208 USA
| | - Susan M Aronica
- Department of Biology, Canisius College, 2001 Main Street, Buffalo, NY 14208 USA
| |
Collapse
|
157
|
Zatula N, Wiese M, Bunzendahl J, Birchmeier W, Perske C, Bleckmann A, Brembeck FH. The BCL9-2 proto-oncogene governs estrogen receptor alpha expression in breast tumorigenesis. Oncotarget 2015; 5:6770-87. [PMID: 25149534 PMCID: PMC4196162 DOI: 10.18632/oncotarget.2252] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The majority of human breast cancers express estrogen receptor alpha (ER), which is important for therapy with anti-estrogens. Here we describe the role of BCL9-2, a proto-oncogene previously characterized as co-activator of Wnt/ß-catenin signaling, for mammary tumorigenesis in mice and human. ER positive human breast cancers showed overexpression of BCL9-2 and tamoxifen treated patients with high BCL9-2 demonstrated a better survival. BCL9-2 was upregulated during puberty and pregnancy in normal mammary epithelia, but downregulated in the involuted gland. BCL9-2 overexpression in vivo delayed the mammary involution and induced alveolar hyperplasia. Moreover, aged BCL9-2 transgenic mice developed ductal-like mammary tumors with high nuclear ER expression. We found, that primary cell cultures of BCL9-2 breast tumors responded to tamoxifen treatment. Moreover, BCL9-2 regulated the expression of ER and the proliferation of human breast cancer cells independently of ß-catenin. Finally, we describe a novel mechanism, how BCL9-2 regulates ER transcription by interaction with Sp1 through the proximal ESR1 gene promoter. In summary, BCL9-2 induces ER positive breast cancers in vivo, regulates ER expression by a novel ß-catenin independent mechanism in breast cancer cells, and might predict the therapy response to tamoxifen treatment.
Collapse
Affiliation(s)
- Nathalie Zatula
- Tumor Biology and Signal Transduction, Georg-August-University Göttingen, Germany. Dept. of Hematology and Medical Oncology, Georg-August-University Göttingen, Germany
| | - Maria Wiese
- Tumor Biology and Signal Transduction, Georg-August-University Göttingen, Germany. Dept. of Hematology and Medical Oncology, Georg-August-University Göttingen, Germany
| | - Jens Bunzendahl
- Tumor Biology and Signal Transduction, Georg-August-University Göttingen, Germany. Dept. of Hematology and Medical Oncology, Georg-August-University Göttingen, Germany
| | | | | | - Annalen Bleckmann
- Dept. of Hematology and Medical Oncology, Georg-August-University Göttingen, Germany
| | - Felix H Brembeck
- Tumor Biology and Signal Transduction, Georg-August-University Göttingen, Germany. Dept. of Hematology and Medical Oncology, Georg-August-University Göttingen, Germany
| |
Collapse
|
158
|
Belkaid A, Duguay SR, Ouellette RJ, Surette ME. 17β-estradiol induces stearoyl-CoA desaturase-1 expression in estrogen receptor-positive breast cancer cells. BMC Cancer 2015; 15:440. [PMID: 26022099 PMCID: PMC4446951 DOI: 10.1186/s12885-015-1452-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 05/19/2015] [Indexed: 12/16/2022] Open
Abstract
Background To sustain cell growth, cancer cells exhibit an altered metabolism characterized by increased lipogenesis. Stearoyl-CoA desaturase-1 (SCD-1) catalyzes the production of monounsaturated fatty acids that are essential for membrane biogenesis, and is required for cell proliferation in many cancer cell types. Although estrogen is required for the proliferation of many estrogen-sensitive breast carcinoma cells, it is also a repressor of SCD-1 expression in liver and adipose. The current study addresses this apparent paradox by investigating the impact of estrogen on SCD-1 expression in estrogen receptor-α-positive breast carcinoma cell lines. Methods MCF-7 and T47D mammary carcinomas cells and immortalized MCF-10A mammary epithelial cells were hormone-starved then treated or not with 17β-estradiol. SCD-1 activity was assessed by measuring cellular monounsaturated/saturated fatty acid (MUFA/SFA) ratios, and SCD-1 expression was measured by qPCR, immunoblot, and immunofluorescence analyses. The role of SCD-1 in cell proliferation was measured following treatment with the SCD-1 inhibitor A959372 and following SCD-1 silencing using siRNA. The involvement of IGF-1R on SCD-1 expression was measured using the IGF-1R antagonist AG1024. The expression of SREBP-1c, a transcription factor that regulates SCD-1, was measured by qPCR, and by immunoblot analyses. Results 17β-estradiol significantly induced cell proliferation and SCD-1 activity in MCF-7 and T47D cells but not MCF-10A cells. Accordingly, 17β-estradiol significantly increased SCD-1 mRNA and protein expression in MCF-7 and T47D cells compared to untreated cells. Treatment of MCF-7 cells with 4-OH tamoxifen or siRNA silencing of estrogen receptor-α largely prevented 17β-estradiol-induced SCD-1 expression. 17β-estradiol increased SREBP-1c expression and induced the mature active 60 kDa form of SREBP-1. The selective SCD-1 inhibitor or siRNA silencing of SCD-1 blocked the 17β-estradiol-induced cell proliferation and increase in cellular MUFA/SFA ratios. IGF-1 also induced SCD-1 expression, but to a lesser extent than 17β-estradiol. The IGF-1R antagonist partially blocked 17β-estradiol-induced cell proliferation and SCD-1 expression, suggesting the impact of 17β-estradiol on SCD-1 expression is partially mediated though IGF-1R signaling. Conclusions This study illustrates for the first time that, in contrast to hepatic and adipose tissue, estrogen induces SCD-1 expression and activity in breast carcinoma cells. These results support SCD-1 as a therapeutic target in estrogen-sensitive breast cancer.
Collapse
Affiliation(s)
- Anissa Belkaid
- Department of Chemistry and Biochemistry, Université de Moncton, 18 Antonine Maillet Ave, Moncton, NB, E1A 3E9, Canada. .,Atlantic Cancer Research Institute, Moncton, NB, Canada.
| | - Sabrina R Duguay
- Department of Chemistry and Biochemistry, Université de Moncton, 18 Antonine Maillet Ave, Moncton, NB, E1A 3E9, Canada.
| | | | - Marc E Surette
- Department of Chemistry and Biochemistry, Université de Moncton, 18 Antonine Maillet Ave, Moncton, NB, E1A 3E9, Canada.
| |
Collapse
|
159
|
Liu X, Gold KA, Dmitrovsky E. The Myb-p300 Interaction Is a Novel Molecular Pharmacologic Target. Mol Cancer Ther 2015; 14:1273-5. [PMID: 25995438 DOI: 10.1158/1535-7163.mct-15-0271] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 04/07/2015] [Indexed: 11/16/2022]
Affiliation(s)
- Xi Liu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kathryn A Gold
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ethan Dmitrovsky
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
160
|
Gee JMW, Nicholson RI, Barrow D, Dutkowski CM, Goddard L, Jordan NJ, McClelland RA, Knowlden JM, Francies HE, Hiscox SE, Hutcheson IR. Antihormone induced compensatory signalling in breast cancer: an adverse event in the development of endocrine resistance. Horm Mol Biol Clin Investig 2015; 5:67-77. [PMID: 25961242 DOI: 10.1515/hmbci.2011.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 01/21/2011] [Indexed: 12/16/2022]
Abstract
Using MCF7 breast cancer cells, it has been shown that antihormones promote expression/activity of oestrogen-repressed tyrosine kinases, notably EGFR, HER2 and Src. These inductive events confer responsiveness to targeted inhibitors (e.g., gefitinib, trastuzumab, saracatinib). We observed that these antihormone-induced phenomena are common to ER+HER2- and ER+HER2+ breast cancer models in vitro, where targeting of EGFR, HER2 or Src alongside antihormone improves antitumour response and delays/prevents endocrine resistance. Such targeted inhibitors also subvert acquired endocrine resistant cells which retain increased EGFR, HER2 and Src (e.g., TAMR and FASR models derived after 6-12 months of tamoxifen or Faslodex treatment). Thus, antihormone-induced tyrosine kinases comprise "compensatory signalling" crucial in limiting maximal initial antihormone response and subsequently driving acquired resistance in vitro. However, despite such convincing preclinical findings from our group and others, clinical trials examining equivalent antigrowth factor strategies have proved relatively disappointing. Our new studies deciphering underlying causes reveal that further antihormone-promoted events could be pivotal in vivo. Firstly, Faslodex induces HER3 and HER4 which sensitise ER+ cells to heregulin, a paracrine growth factor that overcomes endocrine response and diminishes antitumour effect of agents targeting EGFR, HER2 or Src alongside antihormone. Secondly, extended antihormone exposure (experienced by ER+ cells prior to adjuvant clinical relapse) can "reprogramme" the compensatory kinase profile in vitro, hindering candidate antigrowth factor targeting of endocrine resistance. Faslodex resistant cells maintained with this antihormone for 3 years in vitro lose EGFR/HER2 dependency, gaining alternative mitogenic/invasion kinases. Deciphering these previously unrecognised antihormone-induced events could provide superior treatments to control endocrine relapse in the clinic.
Collapse
|
161
|
Dago DN, Scafoglio C, Rinaldi A, Memoli D, Giurato G, Nassa G, Ravo M, Rizzo F, Tarallo R, Weisz A. Estrogen receptor beta impacts hormone-induced alternative mRNA splicing in breast cancer cells. BMC Genomics 2015; 16:367. [PMID: 25956916 PMCID: PMC4424892 DOI: 10.1186/s12864-015-1541-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 04/17/2015] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Estrogens play an important role in breast cancer (BC) development and progression; when the two isoforms of the estrogen receptor (ERα and ERβ) are co-expressed each of them mediate specific effects of these hormones in BC cells. ERβ has been suggested to exert an antagonist role toward the oncogenic activities of ERα, and for this reason it is considered an oncosuppressor. As clinical evidence regarding a prognostic role for this receptor subtype in hormone-responsive BC is still limited and conflicting, more knowledge is required on the biological functions of ERβ in cancer cells. We have previously described the ERβ and ERα interactomes from BC cells, identifying specific and distinct patterns of protein interactions for the two receptors. In particular, we identified factors involved in mRNA splicing and maturation as important components of both ERα and ERβ pathways. Guided by these findings, here we performed RNA sequencing to investigate in depth the differences in the early transcriptional events and RNA splicing patterns induced by estradiol in cells expressing ERα alone or ERα and ERβ. RESULTS Exon skipping was the most abundant splicing event in the post-transcriptional regulation by estradiol. We identified several splicing events induced by ERα alone and by ERα+ERβ, demonstrating for the first time that ERβ significantly affects estrogen-induced splicing in BC cells, as revealed by modification of a subset of ERα-dependent splicing by ERβ, as well as by the presence of splicing isoforms only in ERβ+cells. In particular, we observed that ERβ+BC cell lines exhibited around 2-fold more splicing events than the ERβ- cells. Interestingly, we identified putative direct targets of ERβ-mediated alternative splicing by correlating the genomic locations of ERβ and ERα binding sites with estradiol-induced differential splicing in the corresponding genes. CONCLUSIONS Taken together, these results demonstrate that ERβ significantly affects estrogen-induced early transcription and mRNA splicing in hormone-responsive BC cells, providing novel information on the biological role of ERβ in these tumors.
Collapse
Affiliation(s)
- Dougba Noel Dago
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery, University of Salerno, Via S. Allende, 1, Baronissi, SA, 84081, Italy. .,UFR Sciences Biologiques, Université Peleforo Gon Coulibaly, Korhogo, Ivory Coast.
| | - Claudio Scafoglio
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, USA.
| | - Antonio Rinaldi
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery, University of Salerno, Via S. Allende, 1, Baronissi, SA, 84081, Italy.
| | - Domenico Memoli
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery, University of Salerno, Via S. Allende, 1, Baronissi, SA, 84081, Italy.
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery, University of Salerno, Via S. Allende, 1, Baronissi, SA, 84081, Italy.
| | - Giovanni Nassa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery, University of Salerno, Via S. Allende, 1, Baronissi, SA, 84081, Italy.
| | - Maria Ravo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery, University of Salerno, Via S. Allende, 1, Baronissi, SA, 84081, Italy.
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery, University of Salerno, Via S. Allende, 1, Baronissi, SA, 84081, Italy.
| | - Roberta Tarallo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery, University of Salerno, Via S. Allende, 1, Baronissi, SA, 84081, Italy.
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery, University of Salerno, Via S. Allende, 1, Baronissi, SA, 84081, Italy. .,Molecular Pathology and Medical Genomics, "SS. Giovanni di Dio e Ruggi d'Aragona - Schola Medica Salernitana" Hospital of the University of Salerno, Salerno, Italy.
| |
Collapse
|
162
|
H19 lncRNA mediates 17β-estradiol-induced cell proliferation in MCF-7 breast cancer cells. Oncol Rep 2015; 33:3045-52. [PMID: 25846769 DOI: 10.3892/or.2015.3899] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/10/2015] [Indexed: 11/05/2022] Open
|
163
|
Debeb BG, Smith DL, Li L, Larson R, Xu W, Woodward WA. Differential effect of phosphorylation-defective survivin on radiation response in estrogen receptor-positive and -negative breast cancer. PLoS One 2015; 10:e0120719. [PMID: 25763854 PMCID: PMC4357387 DOI: 10.1371/journal.pone.0120719] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 01/26/2015] [Indexed: 01/18/2023] Open
Abstract
Survivin is a key member of the inhibitor of apoptosis protein family, and is considered a promising therapeutic target due to its universal overexpression in cancers. Survivin is implicated in cellular radiation response through its role in apoptosis, cell division, and DNA damage response. In the present study, analysis of publically available data sets showed that survivin gene expression increased with breast cancer stage (p < 0.00001) and was significantly higher in estrogen receptor-negative cancers as compared to estrogen receptor-positive cancers (p = 9e-46). However, survivin was prognostic in estrogen receptor-positive tumors (p = 0.03) but not in estrogen receptor-negative tumors (p = 0.28). We assessed the effect of a survivin dominant-negative mutant on colony-formation (2D) and mammosphere-formation (3D) efficiency, and radiation response in the estrogen receptor-positive MCF7 and estrogen receptor-negative SUM149 breast cancer cell lines. The colony-formation efficiency was significantly lower in the dominant-negative survivin-transduced cells versus control MCF7 cells (0.42 vs. 0.58, p < 0.01), but it was significantly higher in dominant-negative population versus control-transduced SUM149 cells (0.29 vs. 0.20, p < 0.01). A similar, non-significant, trend in mammosphere-formation efficiency was observed. We compared the radiosensitivity of cells stably expressing dominant-negative survivin with their controls in both cell lines under 2D and 3D culture conditions following exposure to increasing doses of radiation. We found that the dominant-negative populations were radioprotective in MCF7 cells but radiosensitive in SUM149 cells compared to the control-transduced population; further, Taxol was synergistic with the survivin mutant in SUM149 but not MCF7. Our data suggests that survivin modulation influences radiation response differently in estrogen receptor-positive and estrogen receptor-negative breast cancer subtypes, warranting further investigation.
Collapse
Affiliation(s)
- Bisrat G. Debeb
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, United States of America
| | - Daniel L. Smith
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, United States of America
| | - Li Li
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, United States of America
| | - Richard Larson
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, United States of America
| | - Wei Xu
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, United States of America
| | - Wendy A. Woodward
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, United States of America
- * E-mail:
| |
Collapse
|
164
|
Bisphenol-A induces expression of HOXC6, an estrogen-regulated homeobox-containing gene associated with breast cancer. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:697-708. [PMID: 25725483 DOI: 10.1016/j.bbagrm.2015.02.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 02/13/2015] [Accepted: 02/17/2015] [Indexed: 12/31/2022]
Abstract
HOXC6 is a homeobox-containing gene associated with mammary gland development and is overexpressed in variety of cancers including breast and prostate cancers. Here, we have examined the expression of HOXC6 in breast cancer tissue, investigated its transcriptional regulation via estradiol (E2) and bisphenol-A (BPA, an estrogenic endocrine disruptor) in vitro and in vivo. We observed that HOXC6 is differentially over-expressed in breast cancer tissue. E2 induces HOXC6 expression in cultured breast cancer cells and in mammary glands of Sprague Dawley rats. HOXC6 expression is also induced upon exposure to BPA both in vitro and in vivo. Estrogen-receptor-alpha (ERα) and ER-coregulators such as MLL-histone methylases are bound to the HOXC6 promoter upon exposure to E2 or BPA and that resulted in increased histone H3K4-trimethylation, histone acetylation, and recruitment of RNA polymerase II at the HOXC6 promoter. HOXC6 overexpression induces expression of tumor growth factors and facilitates growth 3D-colony formation, indicating its potential roles in tumor growth. Our studies demonstrate that HOXC6, which is a critical player in mammary gland development, is upregulated in multiple cases of breast cancer, and is transcriptionally regulated by E2 and BPA, in vitro and in vivo.
Collapse
|
165
|
Singh K, Munuganti RSN, Leblanc E, Lin YL, Leung E, Lallous N, Butler M, Cherkasov A, Rennie PS. In silico discovery and validation of potent small-molecule inhibitors targeting the activation function 2 site of human oestrogen receptor α. Breast Cancer Res 2015; 17:27. [PMID: 25848700 PMCID: PMC4360945 DOI: 10.1186/s13058-015-0529-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 02/02/2015] [Indexed: 12/21/2022] Open
Abstract
Introduction Current approaches to inhibit oestrogen receptor-alpha (ERα) are focused on targeting its hormone-binding pocket and have limitations. Thus, we propose that inhibitors that bind to a coactivator-binding pocket on ERα, called activation function 2 (AF2), might overcome some of these limitations. Methods In silico virtual screening was used to identify small-molecule ERα AF2 inhibitors. These compounds were screened for inhibition of ERα transcriptional activity using stably transfected T47D-KBluc cell line. A direct physical interaction between the AF2 binders and the ERα protein was measured using biolayer interferometry (BLI) and an ERα coactivator displacement assay. Cell viability was assessed by MTS assay in ERα-positive MCF7 cells, tamoxifen-resistant (TamR) cell lines TamR3 and TamR6, and ERα-negative MDA-MB-453 and HeLa cell lines. In addition, ERα inhibition in TamR cells and the effect of compounds on mRNA and protein expression of oestrogen-dependent genes, pS2, cathepsin D and cell division cycle 2 (CDC2) were determined. Results Fifteen inhibitors from two chemical classes, derivatives of pyrazolidine-3,5-dione and carbohydrazide, were identified. In a series of in vitro assays, VPC-16230 of the carbohydrazide chemical class emerged as a lead ERα AF2 inhibitor that significantly downregulated ERα transcriptional activity (half-maximal inhibitory concentration = 5.81 μM). By directly binding to the ERα protein, as confirmed by BLI, VPC-16230 effectively displaced coactivator peptides from the AF2 pocket, confirming its site-specific action. VPC-16230 selectively suppressed the growth of ERα-positive breast cancer cells. Furthermore, it significantly inhibited ERα mediated transcription in TamR cells. More importantly, it reduced mRNA and protein levels of pS2, cathepsin D and CDC2, validating its ER-directed activity. Conclusion We identified VPC-16230 as an ERα AF2-specific inhibitor that demonstrated promising antiproliferative effects in breast cancer cell lines, including TamR cells. VPC-16230 reduced the expression of ERα-inducible genes, including CDC2, which is involved in cell division. We anticipate that the application of ERα AF2 inhibitors will provide a novel approach that can act as a complementary therapeutic to treat ERα-positive, tamoxifen-resistant and metastatic breast cancers. Electronic supplementary material The online version of this article (doi:10.1186/s13058-015-0529-8) contains supplementary material, which is available to authorized users.
Collapse
|
166
|
Application of metabolomics in drug resistant breast cancer research. Metabolites 2015; 5:100-18. [PMID: 25693144 PMCID: PMC4381292 DOI: 10.3390/metabo5010100] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 08/18/2014] [Accepted: 12/24/2014] [Indexed: 12/15/2022] Open
Abstract
The metabolic profiles of breast cancer cells are different from normal mammary epithelial cells. Breast cancer cells that gain resistance to therapeutic interventions can reprogram their endogenous metabolism in order to adapt and proliferate despite high oxidative stress and hypoxic conditions. Drug resistance in breast cancer, regardless of subgroups, is a major clinical setback. Although recent advances in genomics and proteomics research has given us a glimpse into the heterogeneity that exists even within subgroups, the ability to precisely predict a tumor’s response to therapy remains elusive. Metabolomics as a quantitative, high through put technology offers promise towards devising new strategies to establish predictive, diagnostic and prognostic markers of breast cancer. Along with other “omics” technologies that include genomics, transcriptomics, and proteomics, metabolomics fits into the puzzle of a comprehensive systems biology approach to understand drug resistance in breast cancer. In this review, we highlight the challenges facing successful therapeutic treatment of breast cancer and the innovative approaches that metabolomics offers to better understand drug resistance in cancer.
Collapse
|
167
|
Chang YY, Kuo WH, Hung JH, Lee CY, Lee YH, Chang YC, Lin WC, Shen CY, Huang CS, Hsieh FJ, Lai LC, Tsai MH, Chang KJ, Chuang EY. Deregulated microRNAs in triple-negative breast cancer revealed by deep sequencing. Mol Cancer 2015; 14:36. [PMID: 25888956 PMCID: PMC4351690 DOI: 10.1186/s12943-015-0301-9] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 01/21/2015] [Indexed: 12/21/2022] Open
Abstract
Background MicroRNAs (miRNAs) are short, non-coding RNA molecules that play critical roles in human malignancy. However, the regulatory characteristics of miRNAs in triple-negative breast cancer, a phenotype of breast cancer that does not express the genes for estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2, are still poorly understood. Methods In this study, miRNA expression profiles of 24 triple-negative breast cancers and 14 adjacent normal tissues were analyzed using deep sequencing technology. Expression levels of miRNA reads were normalized with the quantile-quantile scaling method. Deregulated miRNAs in triple-negative breast cancer were identified from the sequencing data using the Student’s t-test. Quantitative reverse transcription PCR validations were carried out to examine miRNA expression levels. Potential target candidates of a miRNA were predicted using published target prediction algorithms. Luciferase reporter assay experiments were performed to verify a putative miRNA-target relationship. Validated molecular targets of the deregulated miRNAs were retrieved from curated databases and their associations with cancer progression were discussed. Results A novel 25-miRNA expression signature was found to effectively distinguish triple-negative breast cancers from surrounding normal tissues in a hierarchical clustering analysis. We documented the evidence of seven polycistronic miRNA clusters preferentially harboring deregulated miRNAs in triple-negative breast cancer. Two of these miRNA clusters (miR-143-145 at 5q32 and miR-497-195 at 17p13.1) were markedly down-regulated in triple-negative breast cancer, while the other five miRNA clusters (miR-17-92 at 13q31.3, miR-183-182 at 7q32.2, miR-200-429 at 1p36.33, miR-301b-130b at 22q11.21, and miR-532-502 at Xp11.23) were up-regulated in triple-negative breast cancer. Moreover, miR-130b-5p from the miR-301b-130b cluster was shown to directly repress the cyclin G2 (CCNG2) gene, a crucial cell cycle regulator, in triple-negative breast cancer cells. Luciferase reporter assays showed that miR-130b-5p-mediated repression of CCNG2 was dependent on the sequence of the 3′-untranslated region. The findings described in this study implicate a miR-130b-5p-CCNG2 axis that may be involved in the malignant progression of triple-negative breast cancer. Conclusions Our work delivers a clear picture of the global miRNA regulatory characteristics in triple-negative breast cancer and extends the current knowledge of microRNA regulatory network. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0301-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yao-Yin Chang
- Department of Electrical Engineering, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan.
| | - Wen-Hung Kuo
- Department of Surgery, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Jui-Hui Hung
- Department of Electrical Engineering, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan. .,Bioinformatics and Biostatistics Core, NTU Center of Genomic Medicine, Taipei, Taiwan.
| | - Chien-Yueh Lee
- Department of Electrical Engineering, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan. .,Bioinformatics and Biostatistics Core, NTU Center of Genomic Medicine, Taipei, Taiwan.
| | - Yung-Hua Lee
- Bioinformatics and Biostatistics Core, NTU Center of Genomic Medicine, Taipei, Taiwan.
| | - Ya-Chu Chang
- Institute of Biotechnology, College of Bio-resources and Agriculture, National Taiwan University, Taipei, Taiwan.
| | - Wen-Chun Lin
- Institute of Biotechnology, College of Bio-resources and Agriculture, National Taiwan University, Taipei, Taiwan.
| | - Cheng-Ying Shen
- Institute of Biotechnology, College of Bio-resources and Agriculture, National Taiwan University, Taipei, Taiwan.
| | - Chiun-Sheng Huang
- Department of Surgery, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Fon-Jou Hsieh
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Liang-Chuan Lai
- Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Mong-Hsun Tsai
- Institute of Biotechnology, College of Bio-resources and Agriculture, National Taiwan University, Taipei, Taiwan.
| | - King-Jen Chang
- Department of Surgery, College of Medicine, National Taiwan University, Taipei, Taiwan. .,Department of Surgery, Cheng Ching General Hospital, Taichung, Taiwan.
| | - Eric Y Chuang
- Department of Electrical Engineering, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan. .,Bioinformatics and Biostatistics Core, NTU Center of Genomic Medicine, Taipei, Taiwan.
| |
Collapse
|
168
|
Qian Y, Yin C, Chen Y, Zhang S, Jiang L, Wang F, Zhao M, Liu S. Estrogen contributes to regulating iron metabolism through governing ferroportin signaling via an estrogen response element. Cell Signal 2015; 27:934-42. [PMID: 25660146 DOI: 10.1016/j.cellsig.2015.01.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 01/31/2015] [Indexed: 12/21/2022]
Abstract
Ferroportin (FPN) is the only known iron exporter in mammalian cells, and is universally expressed in most types of cells. FPN signaling plays a crucial role in maintaining iron homeostasis through governing the level of intracellular iron. Serum iron storage is conversely related with the estrogen level in the female bodies, and women in post-menopause are possibly subjected to iron retention. However, the potential effects of estrogen on iron metabolism are not clearly understood. Here, FPN mRNA transcription in all selected estrogen receptor positive (ER+) cells was significantly reduced upon 17β-estradiol (E2) treatment; and this inhibitory effect could be attenuated by ER antagonist tamoxifen. Likewise, in murine bone marrow-derived macrophages (BMDMs), FPN reduction with elevated intracellular iron (reflected by increased ferritin) was observed in response to E2; however, ferritin level barely responded to E2 in FPN-null BMDMs. The observation of inhibition of FPN mRNA expression was not replicated in ER(-) cells upon E2. A functional estrogen response element (ERE) was identified within the promoter of FPN, and this ERE was responsible for the suppressive effect of E2 on FPN expression. Moreover, ovariectomized (OVX) and sham-operated (SHAM) mice were used to further confirm the in vitro finding. The expression of hepatic FPN was induced in OVX mice, compared to that in the SHAM mice. Taken together, our results demonstrated that estrogen is involved in regulating FPN expression through a functional ERE on its promoter, providing additional insights into a vital role of estrogen in iron metabolism.
Collapse
Affiliation(s)
- Yi Qian
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Biological and Environmental Engineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Chunyang Yin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Yue Chen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin 300211, China
| | - Shuping Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Li Jiang
- Department of Nutrition, School of Public Health, Institute of Nutrition and Food Safety, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Fudi Wang
- Department of Nutrition, School of Public Health, Institute of Nutrition and Food Safety, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, Zhejiang 310027, China
| | - Meirong Zhao
- College of Biological and Environmental Engineering, Zhejiang University of Technology, Hangzhou 310032, China.
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| |
Collapse
|
169
|
Bailey ST, Westerling T, Brown M. Loss of estrogen-regulated microRNA expression increases HER2 signaling and is prognostic of poor outcome in luminal breast cancer. Cancer Res 2015; 75:436-45. [PMID: 25388283 PMCID: PMC4297564 DOI: 10.1158/0008-5472.can-14-1041] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Among the genes regulated by estrogen receptor (ER) are miRNAs that play a role in breast cancer signaling pathways. To determine whether miRNAs are involved in ER-positive breast cancer progression to hormone independence, we profiled the expression of 800 miRNAs in the estrogen-dependent human breast cancer cell line MCF7 and its estrogen-independent derivative MCF7:2A (MCF7:2A) using NanoString. We found 78 miRNAs differentially expressed between the two cell lines, including a cluster comprising let-7c, miR99a, and miR125b, which is encoded in an intron of the long noncoding RNA LINC00478. These miRNAs are ER targets in MCF7 cells, and nearby ER binding and their expression are significantly decreased in MCF7:2A cells. The expression of these miRNAs was interrogated in patient samples profiled in The Cancer Genome Atlas (TCGA). Among luminal tumors, these miRNAs are expressed at higher levels in luminal A versus B tumors. Although their expression is uniformly low in luminal B tumors, they are lost only in a subset of luminal A patients. Interestingly, this subset with low expression of these miRNAs had worse overall survival compared with luminal A patients with high expression. We confirmed that miR125b directly targets HER2 and that let-7c also regulates HER2 protein expression. In addition, HER2 protein expression and activity are negatively correlated with let-7c expression in TCGA. In summary, we identified an ER-regulated miRNA cluster that regulates HER2, is lost with progression to estrogen independence, and may serve as a biomarker of poor outcome in ER(+) luminal A breast cancer patients.
Collapse
Affiliation(s)
- Shannon T Bailey
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Thomas Westerling
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Myles Brown
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
170
|
Bolt MJ, Stossi F, Callison AM, Mancini MG, Dandekar R, Mancini MA. Systems level-based RNAi screening by high content analysis identifies UBR5 as a regulator of estrogen receptor-α protein levels and activity. Oncogene 2015; 34:154-64. [PMID: 24441042 PMCID: PMC4871123 DOI: 10.1038/onc.2013.550] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 10/15/2013] [Accepted: 11/08/2013] [Indexed: 12/17/2022]
Abstract
Estrogen receptor-α (ERα) is a central transcription factor that regulates mammary gland physiology and a key driver in breast cancer. In the present study, we aimed to identify novel modulators of ERα-mediated transcriptional regulation via a custom-built siRNA library screen. This screen was directed against a variety of coregulators, transcription modifiers, signaling molecules and DNA damage response proteins. By utilizing a microscopy-based, multi-end point, estrogen responsive biosensor cell line platform, the primary screen identified a wide range of factors that altered ERα protein levels, chromatin remodeling and mRNA output. We then focused on UBR5, a ubiquitin ligase and known oncogene that modulates ERα protein levels and transcriptional output. Finally, we demonstrated that UBR5 also affects endogenous ERα target genes and E2-mediated cell proliferation in breast cancer cells. In conclusion, our multi-end point RNAi screen identified novel modulators of ERα levels and activity, and provided a robust systems level view of factors involved in mechanisms of nuclear receptor action and pathophysiology. Utilizing a high throughput RNAi screening approach we identified UBR5, a protein commonly amplified in breast cancer, as a novel regulator of ERα protein levels and transcriptional activity.
Collapse
Affiliation(s)
- M J Bolt
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - F Stossi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - A M Callison
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - M G Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - R Dandekar
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - M A Mancini
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
171
|
McDaniel JS, Akula Suresh Babu R, Navarro MM, LeBaron RG. Transcriptional regulation of proteoglycan 4 by 17β-estradiol in immortalized baboon temporomandibular joint disc cells. Eur J Oral Sci 2014; 122:100-8. [PMID: 24621258 DOI: 10.1111/eos.12119] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2014] [Indexed: 01/05/2023]
Abstract
Temporomandibular joint disorders (TMDs) affect a significant portion of the population of the USA, with the majority of those seeking treatment being women of childbearing age. Owing to this striking sexual dimorphism it has been postulated that sex hormones play a role in the maintenance of normal temporomandibular joint (TMJ) function. Proteoglycan 4 (PRG4) is a secreted lubricating molecule required for maintaining low frictional levels within articular joints; however, its role in the TMJ is not well characterized. In this study we describe the development of immortalized baboon cells isolated from specific regions of the TMJ disc and their use in the investigation of PRG4 expression and localization patterns in the TMJ. We identified conserved estrogen response elements within the 5' flanking region of the PRG4 gene of several species, and found that treatment of baboon TMJ disc cells with estrogen led to reduced PRG4 promoter activity and reduced expression of PRG4 mRNA in vitro. The observed negative regulation of PRG4 by estrogen could lead to increased friction and degradation of joint components over time. This study, for the first time, provides evidence of the regulatory potential of estrogen on PRG4 gene expression and suggests a novel etiology for the gender disparity observed among TMD patients.
Collapse
|
172
|
Nyante SJ, Gammon MD, Kaufman JS, Bensen JT, Lin DY, Barnholtz-Sloan JS, Hu Y, He Q, Luo J, Millikan RC. Genetic variation in estrogen and progesterone pathway genes and breast cancer risk: an exploration of tumor subtype-specific effects. Cancer Causes Control 2014; 26:121-31. [PMID: 25421376 DOI: 10.1007/s10552-014-0491-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 11/04/2014] [Indexed: 12/23/2022]
Abstract
PURPOSE To determine whether associations between estrogen pathway-related single nucleotide polymorphisms (SNPs) and breast cancer risk differ by molecular subtype, we evaluated associations between SNPs in cytochrome P450 family 19 subfamily A polypeptide 1 (CYP19A1), estrogen receptor (ESR1), 3-beta hydroxysteroid dehydrogenase type I (HSD3B1), 17-beta hydroxysteroid dehydrogenase type II (HSD17B2), progesterone receptor (PGR), and sex hormone-binding globulin (SHBG) and breast cancer risk in a case-control study in North Carolina. METHODS Cases (n = 1,972) were women 20-74 years old and diagnosed with breast cancer between 1993 and 2001. Population-based controls (n = 1,776) were frequency matched to cases by age and race. A total of 195 SNPs were genotyped, and linkage disequilibrium was evaluated using the r (2) statistic. Odds ratios (ORs) and 95 % confidence intervals (CIs) for associations with breast cancer overall and by molecular subtype were estimated using logistic regression. Monte Carlo methods were used to control for multiple comparisons; two-sided p values <3.3 × 10(-4) were statistically significant. Heterogeneity tests comparing the two most common subtypes, luminal A (n = 679) and basal-like (n = 200), were based on the Wald statistic. RESULTS ESR1 rs6914211 (AA vs. AT+TT, OR 2.24, 95 % CI 1.51-3.33), ESR1 rs985191 (CC vs. AA, OR 2.11, 95 % CI 1.43-3.13), and PGR rs1824128 (TT+GT vs. GG, OR 1.33, 95 % CI 1.14-1.55) were associated with risk after accounting for multiple comparisons. Rs6914211 and rs985191 were in strong linkage disequilibrium among controls (African-Americans r (2) = 0.70; whites r (2) = 0.95). There was no evidence of heterogeneity between luminal A and basal-like subtypes, and the three SNPs were also associated with elevated risk of the less common luminal B, HER2+/ER-, and unclassified subtypes. CONCLUSIONS ESR1 and PGR SNPs were associated with risk, but lack of heterogeneity between subtypes suggests variants in hormone-related genes may play similar roles in the etiology of breast cancer molecular subtypes.
Collapse
Affiliation(s)
- Sarah J Nyante
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Fan P, Cunliffe HE, Griffith OL, Agboke FA, Ramos P, Gray JW, Jordan VC. Identification of gene regulation patterns underlying both oestrogen- and tamoxifen-stimulated cell growth through global gene expression profiling in breast cancer cells. Eur J Cancer 2014; 50:2877-86. [PMID: 25212499 PMCID: PMC4210771 DOI: 10.1016/j.ejca.2014.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/22/2014] [Accepted: 08/07/2014] [Indexed: 01/13/2023]
Abstract
PURPOSE A c-Src inhibitor blocks oestrogen (E2)-induced stress and converts E2 responses from inducing apoptosis to growth stimulation in E2-deprived breast cancer cells. A reprogrammed cell line, MCF-7:PF, results in a functional oestrogen receptor (ER). We addressed the question of whether the selective ER modulator 4-hydroxytamoxifen (4-OHT) could target ER to prevent E2-stimulated growth in MCF-7:PF cells. METHODS Expression of mRNA was measured through real-time RT-PCR. Global gene expression profile was analysed through microarray. Transcriptome profiles were screened by RNA-sequencing. RESULTS Unexpectedly, both 4-OHT and E2 stimulated cell growth in a concentration-dependent manner. Expression profiling showed a remarkable overlap in genes regulated in the same direction by E2 and 4-OHT. Pathway enrichment analysis of the 280 genes commonly deregulated in MCF-7:PF cells by 4-OHT and E2 revealed functions mainly related to membrane, cytoplasm and metabolic processes. Further analysis of 98 genes up-regulated by both 4-OHT and E2 uncovered a significant enrichment in genes associated with membrane remodelling, cytoskeleton reorganisation, cytoplasmic adapter proteins, cytoplasm organelle proteins and related processes. 4-OHT was more potent than E2 in up-regulating some membrane remodelling molecules, such as EHD2, FHL2, HOMER3 and RHOF. In contrast, 4-OHT acted as an antagonist to inhibit expression of the majority of enriched membrane-associated genes in wild-type MCF-7 cells. CONCLUSIONS Long-term selection pressure has changed the cell population responses to 4-OHT. Membrane-associated signalling is critical for 4-OHT-stimulated cell growth in MCF-7:PF cells. This study provides a rationale for the further investigation of target therapy for tamoxifen resistant patients.
Collapse
Affiliation(s)
- Ping Fan
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC 20057, United States
| | - Heather E Cunliffe
- Computational Biology Division, The Translational Genomics Research Institute, Phoenix, AZ 85004, United States
| | - Obi L Griffith
- Department of Medicine, Division of Oncology, The Genome Institute, Washington University, St. Louis, MO 63108, United States
| | - Fadeke A Agboke
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC 20057, United States
| | - Pilar Ramos
- Computational Biology Division, The Translational Genomics Research Institute, Phoenix, AZ 85004, United States
| | - Joe W Gray
- Biomedical Engineering Department, Oregon Health and Science University, Portland, OR 97239, United States
| | - V Craig Jordan
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC 20057, United States.
| |
Collapse
|
174
|
Mansouri-Attia N, James R, Ligon A, Li X, Pangas SA. Soy promotes juvenile granulosa cell tumor development in mice and in the human granulosa cell tumor-derived COV434 cell line. Biol Reprod 2014; 91:100. [PMID: 25165122 PMCID: PMC4435027 DOI: 10.1095/biolreprod.114.120899] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 05/15/2014] [Accepted: 08/07/2014] [Indexed: 12/25/2022] Open
Abstract
Soy attracts attention for its health benefits, such as lowering cholesterol or preventing breast and colon cancer. Soybeans contain isoflavones, which act as phytoestrogens. Even though isoflavones have beneficial health effects, a role for isoflavones in the initiation and progression of diseases including cancer is becoming increasingly recognized. While data from rodent studies suggest that neonatal exposure to genistein (the predominant isoflavone in soy) disrupts normal reproductive function, its role in ovarian cancers, particularly granulosa cell tumors (GCT), is largely unknown. Our study aimed to define the contribution of a soy diet in GCT development using a genetically modified mouse model for juvenile GCTs (JGCT; Smad1 Smad5 conditional double knockout mice) as well as a human JGCT cell line (COV434). While dietary soy cannot initiate JGCT development in mice, we show that it has dramatic effects on GCT growth and tumor progression compared to a soy-free diet. Loss of Smad1 and Smad5 alters estrogen receptor alpha (Esr1) expression in granulosa cells, perhaps sensitizing the cells to the effects of genistein. In addition, we found that genistein modulates estrogen receptor expression in the human JGCT cell line and positively promotes cell growth in part by suppressing caspase-dependent apoptosis. Combined, our work suggests that dietary soy consumption has deleterious effects on GCT development.
Collapse
Affiliation(s)
| | - Rebecca James
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas
| | - Alysse Ligon
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas
| | - Xiaohui Li
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas
| | - Stephanie A Pangas
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
175
|
Andruska N, Zheng X, Yang X, Helferich WG, Shapiro DJ. Anticipatory estrogen activation of the unfolded protein response is linked to cell proliferation and poor survival in estrogen receptor α-positive breast cancer. Oncogene 2014; 34:3760-9. [PMID: 25263449 PMCID: PMC4377305 DOI: 10.1038/onc.2014.292] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 07/11/2014] [Accepted: 08/01/2014] [Indexed: 12/29/2022]
Abstract
In response to cell stress, cancer cells often activate the endoplasmic reticulum (EnR) stress sensor, the unfolded protein response (UPR). Little was known about the potential role in cancer of a different mode of UPR activation; anticipatory activation of the UPR prior to accumulation of unfolded protein or cell stress. We show that estrogen, acting via estrogen receptor α (ERα), induces rapid anticipatory activation of the UPR, resulting in increased production of the antiapoptotic chaperone BiP/GRP78, preparing cancer cells for the increased protein production required for subsequent estrogen-ERα induced cell proliferation. In ERα containing cancer cells, the estrogen, 17β-estradiol (E2) activates the UPR through a phospholipase C γ (PLCγ)-mediated opening of EnR IP3R calcium channels, enabling passage of calcium from the lumen of the EnR into the cytosol. siRNA knockdown of ERα blocked the estrogen-mediated increase in cytosol calcium and UPR activation. Knockdown or inhibition of PLCγ, or of IP3R, strongly inhibited the estrogen-mediated increases in cytosol calcium, UPR activation and cell proliferation. E2-ERα activates all three arms of the UPR in breast and ovarian cancer cells in culture and in a mouse xenograft. Knockdown of ATF6α, which regulates UPR chaperones, blocked estrogen induction of BiP and strongly inhibited E2-ERα stimulated cell proliferation. Mild and transient UPR activation by estrogen promotes an adaptive UPR response that protects cells against subsequent UPR-mediated apoptosis. Analysis of data from ERα positive breast cancers demonstrates elevated expression of a UPR gene signature that is a powerful new prognostic marker tightly correlated with subsequent resistance to tamoxifen therapy, reduced time to recurrence and poor survival. Thus, as an early component of the E2-ERα proliferation program, the mitogen estrogen, drives rapid anticipatory activation of the UPR. Anticipatory activation of the UPR is a new role for estrogens in cancer cell proliferation and resistance to therapy.
Collapse
Affiliation(s)
- N Andruska
- 1] Department of Biochemistry, University of Illinois, Urbana, IL, USA [2] College of Medicine, University of Illinois, Urbana, IL, USA
| | - X Zheng
- Department of Biochemistry, University of Illinois, Urbana, IL, USA
| | - X Yang
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, USA
| | - W G Helferich
- 1] College of Medicine, University of Illinois, Urbana, IL, USA [2] Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, USA [3] University of Illinois Cancer Center, Urbana, IL, USA
| | - D J Shapiro
- 1] Department of Biochemistry, University of Illinois, Urbana, IL, USA [2] College of Medicine, University of Illinois, Urbana, IL, USA [3] University of Illinois Cancer Center, Urbana, IL, USA
| |
Collapse
|
176
|
Gong P, Madak-Erdogan Z, Li J, Cheng J, Greenlief CM, Helferich W, Katzenellenbogen JA, Katzenellenbogen BS. Transcriptomic analysis identifies gene networks regulated by estrogen receptor α (ERα) and ERβ that control distinct effects of different botanical estrogens. NUCLEAR RECEPTOR SIGNALING 2014; 12:e001. [PMID: 25363786 PMCID: PMC4193135 DOI: 10.1621/nrs.12001] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 04/28/2014] [Accepted: 05/13/2014] [Indexed: 12/31/2022]
Abstract
The estrogen receptors (ERs) ERα and ERβ mediate the actions of endogenous estrogens as well as those of botanical estrogens (BEs) present in plants. BEs are ingested in the diet and also widely consumed by postmenopausal women as dietary supplements, often as a substitute for the loss of endogenous estrogens at menopause. However, their activities and efficacies, and similarities and differences in gene expression programs with respect to endogenous estrogens such as estradiol (E2) are not fully understood. Because gene expression patterns underlie and control the broad physiological effects of estrogens, we have investigated and compared the gene networks that are regulated by different BEs and by E2. Our aim was to determine if the soy and licorice BEs control similar or different gene expression programs and to compare their gene regulations with that of E2. Gene expression was examined by RNA-Seq in human breast cancer (MCF7) cells treated with control vehicle, BE or E2. These cells contained three different complements of ERs, ERα only, ERα+ERβ, or ERβ only, reflecting the different ratios of these two receptors in different human breast cancers and in different estrogen target cells. Using principal component, hierarchical clustering, and gene ontology and interactome analyses, we found that BEs regulated many of the same genes as did E2. The genes regulated by each BE, however, were somewhat different from one another, with some genes being regulated uniquely by each compound. The overlap with E2 in regulated genes was greatest for the soy isoflavones genistein and S-equol, while the greatest difference from E2 in gene expression pattern was observed for the licorice root BE liquiritigenin. The gene expression pattern of each ligand depended greatly on the cell background of ERs present. Despite similarities in gene expression pattern with E2, the BEs were generally less stimulatory of genes promoting proliferation and were more pro-apoptotic in their gene regulations than E2. The distinctive patterns of gene regulation by the individual BEs and E2 may underlie differences in the activities of these soy and licorice-derived BEs in estrogen target cells containing different levels of the two ERs.
Collapse
Affiliation(s)
| | | | - Jilong Li
- Botanical Research Center, University of Missouri, Columbia, MO 65211
| | - Jianlin Cheng
- Botanical Research Center, University of Missouri, Columbia, MO 65211
| | | | | | | | | |
Collapse
|
177
|
Xu WG, Shang YL, Cong XR, Bian X, Yuan Z. MicroRNA-135b promotes proliferation, invasion and migration of osteosarcoma cells by degrading myocardin. Int J Oncol 2014; 45:2024-32. [PMID: 25190111 DOI: 10.3892/ijo.2014.2641] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/23/2014] [Indexed: 11/06/2022] Open
Abstract
MicroRNA-135b (miR‑135b) functions as an oncogene in malignant tumors suggesting that it might be a target for malignant tumor therapy, but its role in osteosarcoma has not been investigated to date. In this study, we found that myocardin expression is specifically attenuated in MG63 osteosarcoma cells. Silencing miR‑135b restores the expression of myocardin and miR‑135b inhibits the expression of myocardin by targeting its 3'UTR in MG63 osteosarcoma cells. In additional experiments, we found that miR‑135b was highly expressed in osteosarcoma tissues and it promoted MG63 cell proliferation, migration and invasion. Myocardin had the opposite effects of miR‑135b, which suppressed proliferation, migration and invasion in MG63 cells. Thus, it is probably that miR‑135b promotes proliferation, invasion and migration of osteosarcoma cells by degrading myocardin.
Collapse
Affiliation(s)
- Wei-Guo Xu
- Department of Orthopedics, Shanxian County Dongda Hospital, Shanxian, Heze, Shandong 274300, P.R. China
| | - Ying-Lie Shang
- Department of Orthopedics, Qianfoshan Hospital Affiliated to Shandong University, Jinan 250014, P.R. China
| | - Xian-Rui Cong
- Department of Orthopedics, Shanxian County Dongda Hospital, Shanxian, Heze, Shandong 274300, P.R. China
| | - Xia Bian
- Department of Oncology, The People's Hospital of Dongying, East District, Dongying, Shandong 257091, P.R. China
| | - Zhen Yuan
- Department of Orthopedics, The 2nd People's Hospital of Dongying, Dawang Town, Guangrao, Dongying, Shandong 257335, P.R. China
| |
Collapse
|
178
|
Liu X, An BH, Kim MJ, Park JH, Kang YS, Chang M. Human glutathione S-transferase P1-1 functions as an estrogen receptor α signaling modulator. Biochem Biophys Res Commun 2014; 452:840-4. [DOI: 10.1016/j.bbrc.2014.09.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 09/03/2014] [Indexed: 11/17/2022]
|
179
|
Al-Zaubai N, Johnstone CN, Leong MM, Li J, Rizzacasa M, Stewart AG. Resolvin D2 supports MCF-7 cell proliferation via activation of estrogen receptor. J Pharmacol Exp Ther 2014; 351:172-80. [PMID: 25077525 DOI: 10.1124/jpet.114.214403] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Inflammation has been implicated in tumor initiation, angiogenesis, and metastasis, and linked to the development of more aggressive, therapy-resistant estrogen receptor (ER)-positive breast cancer. Resolvin D2 (RvD2) is a potent anti-inflammatory lipid mediator. As RvD2 may be synthesized within breast tumors by both tumor cells and the surrounding stroma cells and is present in plasma at bioactive concentrations, we sought to characterize the impact of RvD2 on cell processes underlying breast tumor growth and spread. Trypan-blue exclusion, transfection with estrogen response element (ERE) reporter, real-time quantitative polymerase chain reaction, competitive radioligand binding assays, Western blotting, and immunofluorescence were the techniques used. Unexpectedly, whereas RvD2 (10-1000 nM) supported the proliferation of the ER-positive breast tumor (MCF-7) cells, it did not affect the ER-negative MDA-MB-231 cell number. The proliferative effect of RvD2 in MCF-7 cells was attenuated by the ER antagonist ICI 182,780 (7α-[9-[(4,4,5,5,5-pentafluoropentyl)sulfinyl]nonyl]estra-1,3,5(10)-triene-3,17β-diol). Furthermore, RvD2 increased ERE transcriptional activity in a number of ER-positive breast and ovarian tumor cell lines. This activation was also inhibited by ICI 182,780. RvD2 altered the expression of a subset of estrogen-responsive genes. Although binding experiments showed that RvD2 did not directly compete with [(3)H]17β-estradiol for ER binding, prior exposure of MCF-7 cells to RvD2 resulted in a significant reduction in the apparent cytosolic ER density. Confocal immunocytochemistry and Western blotting studies showed that RvD2 promoted nuclear localization of ERα. These observations indicate that RvD2 displays significant but indirect estrogenic properties and has the potential to play a role in estrogen-dependent breast cancer progression.
Collapse
Affiliation(s)
- Nuha Al-Zaubai
- Department of Pharmacology and Therapeutics (N.A.-Z., A.G.S.), Peter MacCallum Cancer Centre (C.N.J.), School of Chemistry, the Bio21 Institute (M.M.L., J.L., M.R.), Sir Peter MacCallum Department of Oncology (C.N.J.), and Department of Pathology (C.N.J.), University of Melbourne, Victoria, Australia
| | - Cameron N Johnstone
- Department of Pharmacology and Therapeutics (N.A.-Z., A.G.S.), Peter MacCallum Cancer Centre (C.N.J.), School of Chemistry, the Bio21 Institute (M.M.L., J.L., M.R.), Sir Peter MacCallum Department of Oncology (C.N.J.), and Department of Pathology (C.N.J.), University of Melbourne, Victoria, Australia
| | - May May Leong
- Department of Pharmacology and Therapeutics (N.A.-Z., A.G.S.), Peter MacCallum Cancer Centre (C.N.J.), School of Chemistry, the Bio21 Institute (M.M.L., J.L., M.R.), Sir Peter MacCallum Department of Oncology (C.N.J.), and Department of Pathology (C.N.J.), University of Melbourne, Victoria, Australia
| | - John Li
- Department of Pharmacology and Therapeutics (N.A.-Z., A.G.S.), Peter MacCallum Cancer Centre (C.N.J.), School of Chemistry, the Bio21 Institute (M.M.L., J.L., M.R.), Sir Peter MacCallum Department of Oncology (C.N.J.), and Department of Pathology (C.N.J.), University of Melbourne, Victoria, Australia
| | - Mark Rizzacasa
- Department of Pharmacology and Therapeutics (N.A.-Z., A.G.S.), Peter MacCallum Cancer Centre (C.N.J.), School of Chemistry, the Bio21 Institute (M.M.L., J.L., M.R.), Sir Peter MacCallum Department of Oncology (C.N.J.), and Department of Pathology (C.N.J.), University of Melbourne, Victoria, Australia
| | - Alastair G Stewart
- Department of Pharmacology and Therapeutics (N.A.-Z., A.G.S.), Peter MacCallum Cancer Centre (C.N.J.), School of Chemistry, the Bio21 Institute (M.M.L., J.L., M.R.), Sir Peter MacCallum Department of Oncology (C.N.J.), and Department of Pathology (C.N.J.), University of Melbourne, Victoria, Australia
| |
Collapse
|
180
|
|
181
|
Fouz N, Amid A, Hashim YZHY. Gene Expression Analysis in MCF-7 Breast Cancer Cells Treated with Recombinant Bromelain. Appl Biochem Biotechnol 2014; 173:1618-39. [DOI: 10.1007/s12010-014-0947-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 04/23/2014] [Indexed: 10/25/2022]
|
182
|
Periodic Estrogen Receptor-Beta Activation: A Novel Approach to Prevent Ischemic Brain Damage. Neurochem Res 2014; 40:2009-17. [PMID: 24906488 DOI: 10.1007/s11064-014-1346-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 05/12/2014] [Accepted: 05/22/2014] [Indexed: 02/06/2023]
Abstract
In women, the risk for cerebral ischemia climbs rapidly after menopause. At menopause, production of ovarian hormones; i.e., progesterone and estrogen, slowly diminishes. Estrogen has been suggested to confer natural protection to premenopausal women from ischemic stroke and some of its debilitating consequences. This notion is also strongly supported by laboratory studies showing that a continuous chronic 17β-estradiol (E2; a potent estrogen) regimen protects brain from ischemic injury. However, concerns regarding the safety of the continuous intake of E2 were raised by the failed translation to the clinic. Recent studies demonstrated that repetitive periodic E2 pretreatments, in contrast to continuous E2 treatment, provided neuroprotection against cerebral ischemia in ovariectomized rats. Periodic E2 pretreatment protects hippocampal neurons through activation of estrogen receptor subtype beta (ER-β). Apart from neuroprotection, periodic activation of ER-β in ovariectomized rats significantly improves hippocampus-dependent learning and memory. Difficulties in learning and memory loss are the major consequence of ischemic brain damage. Periodic ER-β agonist pretreatment may provide pharmacological access to a protective state against ischemic stroke and its debilitating consequences. The use of ER-β-selective agonists constitutes a safer target for future research than ER-α agonist or E2, inasmuch as it lacks the ability to stimulate the proliferation of breast or endometrial tissue. In this review, we highlight ER-β signaling as a guide for future translational research to reduce cognitive decline and cerebral ischemia incidents/impact in post-menopausal women, while avoiding the side effects produced by chronic E2 treatment.
Collapse
|
183
|
Haricharan S, Bainbridge MN, Scheet P, Brown PH. Somatic mutation load of estrogen receptor-positive breast tumors predicts overall survival: an analysis of genome sequence data. Breast Cancer Res Treat 2014; 146:211-20. [PMID: 24839032 PMCID: PMC4061465 DOI: 10.1007/s10549-014-2991-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 04/30/2014] [Indexed: 01/03/2023]
Abstract
Breast cancer is one of the most commonly diagnosed cancers in women. While there are several effective therapies for breast cancer and important single gene prognostic/predictive markers, more than 40,000 women die from this disease every year. The increasing availability of large-scale genomic datasets provides opportunities for identifying factors that influence breast cancer survival in smaller, well-defined subsets. The purpose of this study was to investigate the genomic landscape of various breast cancer subtypes and its potential associations with clinical outcomes. We used statistical analysis of sequence data generated by the Cancer Genome Atlas initiative including somatic mutation load (SML) analysis, Kaplan–Meier survival curves, gene mutational frequency, and mutational enrichment evaluation to study the genomic landscape of breast cancer. We show that ER+, but not ER−, tumors with high SML associate with poor overall survival (HR = 2.02). Further, these high mutation load tumors are enriched for coincident mutations in both DNA damage repair and ER signature genes. While it is known that somatic mutations in specific genes affect breast cancer survival, this study is the first to identify that SML may constitute an important global signature for a subset of ER+ tumors prone to high mortality. Moreover, although somatic mutations in individual DNA damage genes affect clinical outcome, our results indicate that coincident mutations in DNA damage response and signature ER genes may prove more informative for ER+ breast cancer survival. Next generation sequencing may prove an essential tool for identifying pathways underlying poor outcomes and for tailoring therapeutic strategies.
Collapse
Affiliation(s)
- Svasti Haricharan
- Department of Clinical Cancer Prevention, Unit 1360, The University of Texas M.D. Anderson Cancer Center, P.O. Box 301439, Houston, TX, 77030-1439, USA,
| | | | | | | |
Collapse
|
184
|
Viedma-Rodríguez R, Baiza-Gutman L, Salamanca-Gómez F, Diaz-Zaragoza M, Martínez-Hernández G, Ruiz Esparza-Garrido R, Velázquez-Flores MA, Arenas-Aranda D. Mechanisms associated with resistance to tamoxifen in estrogen receptor-positive breast cancer (review). Oncol Rep 2014; 32:3-15. [PMID: 24841429 DOI: 10.3892/or.2014.3190] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 04/03/2014] [Indexed: 11/06/2022] Open
Abstract
Anti-estrogens such as tamoxifen are widely used in the clinic to treat estrogen receptor-positive breast tumors. Patients with estrogen receptor-positive breast cancer initially respond to treatment with anti-hormonal agents such as tamoxifen, but remissions are often followed by the acquisition of resistance and, ultimately, disease relapse. The development of a rationale for the effective treatment of tamoxifen-resistant breast cancer requires an understanding of the complex signal transduction mechanisms. In the present study, we explored some mechanisms associated with resistance to tamoxifen, such as pharmacologic mechanisms, loss or modification in estrogen receptor expression, alterations in co-regulatory proteins and the regulation of the different signaling pathways that participate in different cellular processes such as survival, proliferation, stress, cell cycle, inhibition of apoptosis regulated by the Bcl-2 family, autophagy, altered expression of microRNA, and signaling pathways that regulate the epithelial-mesenchymal transition in the tumor microenvironment. Delineation of the molecular mechanisms underlying the development of resistance may aid in the development of treatment strategies to enhance response and compromise resistance.
Collapse
Affiliation(s)
- Rubí Viedma-Rodríguez
- Molecular Genetics Laboratory, Medical Research Unit in Human Genetics, Pediatric Hospital, National Medical Center Century XXI (CMN-SXXI), Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | - Luis Baiza-Gutman
- Unit of Morphology and Function, Faculty of Higher Studies (FES) Iztacala, National Autonomous University of Mexico (UNAM), Los Reyes Iztacala, State of Mexico, Mexico
| | - Fabio Salamanca-Gómez
- Molecular Genetics Laboratory, Medical Research Unit in Human Genetics, Pediatric Hospital, National Medical Center Century XXI (CMN-SXXI), Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | | | - Guadalupe Martínez-Hernández
- Unit of Morphology and Function, Faculty of Higher Studies (FES) Iztacala, National Autonomous University of Mexico (UNAM), Los Reyes Iztacala, State of Mexico, Mexico
| | - Ruth Ruiz Esparza-Garrido
- Molecular Genetics Laboratory, Medical Research Unit in Human Genetics, Pediatric Hospital, National Medical Center Century XXI (CMN-SXXI), Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | - Miguel Angel Velázquez-Flores
- Molecular Genetics Laboratory, Medical Research Unit in Human Genetics, Pediatric Hospital, National Medical Center Century XXI (CMN-SXXI), Mexican Social Security Institute (IMSS), Mexico City, Mexico
| | - Diego Arenas-Aranda
- Molecular Genetics Laboratory, Medical Research Unit in Human Genetics, Pediatric Hospital, National Medical Center Century XXI (CMN-SXXI), Mexican Social Security Institute (IMSS), Mexico City, Mexico
| |
Collapse
|
185
|
Shi JA, Lu DL, Huang X, Tan W. miR-219 inhibits the proliferation, migration and invasion of medulloblastoma cells by targeting CD164. Int J Mol Med 2014; 34:237-43. [PMID: 24756834 DOI: 10.3892/ijmm.2014.1749] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 04/01/2014] [Indexed: 02/05/2023] Open
Abstract
It is known that microRNA-219 (miR-219) expression is downregulated in medulloblastoma. In the present study, we investigated the expression, targets and functional effects of miR-219 in D283-MED medulloblastoma cells. We first demonstrated that miR-219 not only inhibits proliferation, but also suppresses the invasion and migration of D283-MED cells. Moreover, the knockdown of miR-219 promoted the proliferation, migration and invasion of the D283-MED cells. Secondly, we predicted that miR-219 targets the 3' untranslated region (3'UTR) of CD164 and orthodenticle homeobox 2 (OTX2) and then confirmed that it significantly downregulated the protein expression of CD164 and OTX2 in D283-MED cells. Finally, we demonstrated that the proliferation, invasion and migration of D283-MED cells were promoted by theectopic expression of CD164. These results indicate that miR-219 suppresses the proliferation, migration and invasion of medulloblastoma cells by targeting CD164. The results also suggest that miR-219 may serve as a potential therapeutic agent for medulloblastoma.
Collapse
Affiliation(s)
- Jia-Ang Shi
- Department of Psychiatry, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Da-Lin Lu
- Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei 430081, P.R. China
| | - Xuan Huang
- Medical College, Nanchang University, Jiangxi 330000, P.R. China
| | - Wei Tan
- Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei 430081, P.R. China
| |
Collapse
|
186
|
Jonsson P, Katchy A, Williams C. Support of a bi-faceted role of estrogen receptor β (ERβ) in ERα-positive breast cancer cells. Endocr Relat Cancer 2014; 21:143-60. [PMID: 24192230 PMCID: PMC3946733 DOI: 10.1530/erc-13-0444] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The expression of estrogen receptor α (ERα) in breast cancer identifies patients most likely to respond to endocrine treatment. The second ER, ERβ, is also expressed in breast tumors, but its function and therapeutic potential need further study. Although in vitro studies have established that ERβ opposes transcriptional and proliferative functions of ERα, several clinical studies report its correlation with proliferative markers and poorer prognosis. The data demonstrate that ERβ opposes ERα are primarily based on transient expression of ERβ. Here, we explored the functions of constitutively expressed ERβ in ERα-positive breast cancer lines MCF7 and T47D. We found that ERβ, under these conditions heterodimerized with ERα in the presence and absence of 17β-estradiol, and induced genome-wide transcriptional changes. Widespread anti-ERα signaling was, however, not observed and ERβ was not antiproliferative. Tamoxifen antagonized proliferation and ER-mediated gene regulation both in the presence and absence of ERβ. In conclusion, ERβ's role in cells adapted to its expression appears to differ from its role in cells with transient expression. Our study is important because it provides a deeper understanding of ERβ's role in breast tumors that coexpress both receptors and supports an emerging bi-faceted role of ERβ.
Collapse
Affiliation(s)
| | | | - Cecilia Williams
- To whom correspondence should be addressed:, Postal address: Center for Nuclear Receptors and Cell Signaling, 3605 Cullen Blvd., SERC Bldg. 545, Houston, TX 77204-5056,
| |
Collapse
|
187
|
Won YS, Lee JH, Kwon SJ, Kim JY, Park KH, Lee MK, Seo KI. α-Mangostin-induced apoptosis is mediated by estrogen receptor α in human breast cancer cells. Food Chem Toxicol 2014; 66:158-65. [DOI: 10.1016/j.fct.2014.01.040] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 01/20/2014] [Accepted: 01/22/2014] [Indexed: 12/13/2022]
|
188
|
Lata K, Mukherjee TK. Knockdown of receptor for advanced glycation end products attenuate 17α-ethinyl-estradiol dependent proliferation and survival of MCF-7 breast cancer cells. Biochim Biophys Acta Gen Subj 2014; 1840:1083-91. [DOI: 10.1016/j.bbagen.2013.11.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/25/2013] [Accepted: 11/09/2013] [Indexed: 12/11/2022]
|
189
|
Candelaria NR, Liu K, Lin CY. Estrogen receptor alpha: molecular mechanisms and emerging insights. J Cell Biochem 2014; 114:2203-8. [PMID: 23649536 DOI: 10.1002/jcb.24584] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 04/29/2013] [Indexed: 11/11/2022]
Abstract
Estrogen receptor alpha (ERα) is a cellular receptor for the female sex hormone estrogen and other natural and synthetic ligands and play critical roles in normal development and physiology and in the etiology and treatment of endocrine-related diseases. ERα is a member of the nuclear receptor superfamily of transcription factors and regulates target gene expression in a ligand-dependent manner. It has also been shown to interact with G-protein coupled receptors and associated signaling molecules in the cytoplasm. Transcriptionally, ERα either binds DNA directly through conserved estrogen response element sequence motifs or indirectly by tethering to other interacting transcription factors and nucleate transcriptional regulatory complexes which include an array of co-regulator proteins. Genome-scale studies of ERα transcriptional activity and localization have revealed mechanistic complexity and insights including novel interactions with several transcription factors, including FOXA1, AP-2g, GATA3, and RUNX1, which function as pioneering, collaborative, or tethering factors. The major challenge and exciting prospect moving forward is the comprehensive definition and integration of ERα complexes and mechanisms and their tissue-specific roles in normal physiology and in human diseases.
Collapse
Affiliation(s)
- Nicholes R Candelaria
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, Texas 77204‐5506, USA
| | | | | |
Collapse
|
190
|
Mir M, Bergamaschi A, Katzenellenbogen BS, Popescu G. Highly sensitive quantitative imaging for monitoring single cancer cell growth kinetics and drug response. PLoS One 2014; 9:e89000. [PMID: 24558461 PMCID: PMC3928317 DOI: 10.1371/journal.pone.0089000] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 01/13/2014] [Indexed: 12/18/2022] Open
Abstract
The detection and treatment of cancer has advanced significantly in the past several decades, with important improvements in our understanding of the fundamental molecular and genetic basis of the disease. Despite these advancements, drug-screening methodologies have remained essentially unchanged since the introduction of the in vitro human cell line screen in 1990. Although the existing methods provide information on the overall effects of compounds on cell viability, they are restricted by bulk measurements, large sample sizes, and lack capability to measure proliferation kinetics at the individual cell level. To truly understand the nature of cancer cell proliferation and to develop personalized adjuvant therapies, there is a need for new methodologies that provide quantitative information to monitor the effect of drugs on cell growth as well as morphological and phenotypic changes at the single cell level. Here we show that a quantitative phase imaging modality known as spatial light interference microscopy (SLIM) addresses these needs and provides additional advantages over existing proliferation assays. We demonstrate these capabilities through measurements on the effects of the hormone estradiol and the antiestrogen ICI182,780 (Faslodex) on the growth of MCF-7 breast cancer cells. Along with providing information on changes in the overall growth, SLIM provides additional biologically relevant information. For example, we find that exposure to estradiol results in rapidly growing cells with lower dry mass than the control population. Subsequently blocking the estrogen receptor with ICI results in slower growing cells, with lower dry masses than the control. This ability to measure changes in growth kinetics in response to environmental conditions provides new insight on growth regulation mechanisms. Our results establish the capabilities of SLIM as an advanced drug screening technology that provides information on changes in proliferation kinetics at the cellular level with greater sensitivity than any existing method.
Collapse
Affiliation(s)
- Mustafa Mir
- Quantitative Light Imaging Laboratory, Department of Electrical and Computer Engineering, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Anna Bergamaschi
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Benita S. Katzenellenbogen
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Gabriel Popescu
- Quantitative Light Imaging Laboratory, Department of Electrical and Computer Engineering, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
191
|
Du Y, Shi A, Han B, Li S, Wu D, Jia H, Zheng C, Ren L, Fan Z. COX-2 silencing enhances tamoxifen antitumor activity in breast cancer in vivo and in vitro. Int J Oncol 2014; 44:1385-93. [PMID: 24535190 DOI: 10.3892/ijo.2014.2299] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 01/16/2014] [Indexed: 11/06/2022] Open
Abstract
Tamoxifen (Tam), a selective estrogen receptor modulator, is in wide clinical use for the treatment and prevention of breast cancer. However, extended TAM administration for breast cancer induces increased VEGF levels in patients, promoting new blood vessel formation and thereby limiting its efficacy and highlighting the need for improved therapeutic strategies. Cyclooxygenase-2 (COX-2) silencing via a replication-incompetent lentivirus (LV-COX-2) induce cancer apoptosis and suppresses VEGF gene expression. In this study, the effect of LV-COX-2 infection, either alone or in combination with TAM, was analyzed in a breast cell lines for suppressing VEGF expression and simultaneously reducing doses of TAM. Cell proliferation, apoptosis, angiogenesis, metastasis, cell cycle distribution, an receptor signaling were determined after LV-COX-2 combination with TAM treatment. In addition, tumor growth ability in nude mice was detected to define the combination treatment effect in tumorigenesis in vivo. It is found that LV-COX-2 combination with TAM treatment in breast cancer cell significantly suppressed the proliferation and metastasis, and induced tumor apoptosis in vitro, and tumor growth also was suppressed in vivo. In addition, we also found that LV-COX-2 combination with TAM treatment could inhibit angiogenesis and VEGF expression. Taken together, our experimental results indicate that LV-COX-2 combination with TAM has promising outcome in anti-metastatic and apoptotic studies. Furthermore, these results showed that LV-COX-2 combination with TAM is a potential drug candidate for treatment of breast tumors expressing high levels of VEGF.
Collapse
Affiliation(s)
- Ye Du
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun 130021, Jilin, P.R. China
| | - Aiping Shi
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun 130021, Jilin, P.R. China
| | - Bing Han
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun 130021, Jilin, P.R. China
| | - Sijie Li
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun 130021, Jilin, P.R. China
| | - Di Wu
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun 130021, Jilin, P.R. China
| | - Hongyao Jia
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun 130021, Jilin, P.R. China
| | - Chao Zheng
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun 130021, Jilin, P.R. China
| | - Liqun Ren
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun 130021, Jilin, P.R. China
| | - Zhimin Fan
- Department of Breast Surgery, The First Hospital, Jilin University, Changchun 130021, Jilin, P.R. China
| |
Collapse
|
192
|
Madak-Erdogan Z, Ventrella R, Petry L, Katzenellenbogen BS. Novel roles for ERK5 and cofilin as critical mediators linking ERα-driven transcription, actin reorganization, and invasiveness in breast cancer. Mol Cancer Res 2014; 12:714-27. [PMID: 24505128 DOI: 10.1158/1541-7786.mcr-13-0588] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
UNLABELLED Cancer cell motility and invasiveness are fundamental characteristics of the malignant phenotype and are regulated through diverse signaling networks involving kinases and transcription factors. This study establishes an estrogen receptor (ERα)/MAPK (ERK5)/cofilin (CFL1) network that specifies the degree of breast cancer cell aggressiveness through coupling of actin reorganization and hormone receptor-mediated transcription. Using dominant negative and constitutively active forms, as well as small-molecule inhibitors of extracellular signal-regulated kinase (ERK)5 and MAP-ERK kinase (MEK)5, it was revealed that hormone activation of ERα determined the subcellular localization of ERK5, which functions as a coregulator of ERα-dependent gene transcription. Notably, ERK5 acted in concert with the actin remodeling protein, CFL1, and upon hormone exposure, both localized to active nuclear transcriptional hubs as verified by immunofluorescence and proximity ligation assays. Both ERK5 and CFL1 facilitated PAF1 recruitment to the RNA Pol II complex and both were required for regulation of gene transcription. In contrast, in cells lacking ERα, ERK5 and CFL1 localized to cytoplasmic membrane regions of high actin remodeling, promoting cell motility and invasion, thereby revealing a mechanism likely contributing to the generally poorer prognosis of patients with ERα-negative breast cancer. Thus, this study uncovers the dynamic interplay of nuclear receptor-mediated transcription and actin reorganization in phenotypes of breast cancer aggressiveness. IMPLICATIONS Identification of the ER/ERK5/CFL1 axis suggests new prognostic biomarkers and novel therapeutic avenues to moderate cancer aggressiveness.
Collapse
Affiliation(s)
- Zeynep Madak-Erdogan
- Authors' Affiliation: Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | | | | | | |
Collapse
|
193
|
Hahm ER, Singh SV. Diallyl trisulfide inhibits estrogen receptor-α activity in human breast cancer cells. Breast Cancer Res Treat 2014; 144:47-57. [PMID: 24487688 DOI: 10.1007/s10549-014-2841-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 01/15/2014] [Indexed: 11/26/2022]
Abstract
Organosulfur compounds from garlic effectively inhibit growth of transplanted as well as spontaneous cancers in preclinical animal models without any adverse side effects. However, the mechanisms underlying anticancer effect of this class of compounds are not fully understood. This study reports, for the first time, that garlic organosulfide diallyl trisulfide (DATS) inhibits estrogen receptor-α (ER-α) activity in human breast cancer cells. Exposure of MCF-7 and T47D cells to DATS resulted in downregulation of ER-α protein, which peaked between 12- and 24-h post-treatment. DATS was relatively more effective in suppressing ER-α protein expression compared with its mono and disulfide analogs. The 17β-estradiol (E2)-induced expression of pS2 and cyclin D1, ER-α target gene products, was also decreased in the presence of DATS. Downregulation of ER-α protein expression resulting from DATS treatment was accompanied by a decrease in nuclear levels of ER-α protein, ER-α mRNA suppression, and inhibition of ERE2e1b-luciferase reporter activity. DATS-mediated inhibition of cell viability and apoptosis induction were not affected in the presence of E2. In agreement with these results, ectopic expression of ER-α in MDA-MB-231 cell line failed to confer any protection against cell proliferation inhibition or apoptosis induction resulting from DATS exposure. DATS treatment caused a decrease in protein levels of peptidyl-prolyl cis-trans isomerase (Pin1), and overexpression of Pin1 partially attenuated ER-α downregulation by DATS. DATS-induced apoptosis was modestly but significantly augmented by overexpression of Pin1. In conclusion, this study identifies ER-α as a novel target of DATS in mammary cancer cells.
Collapse
Affiliation(s)
- Eun-Ryeong Hahm
- Department of Pharmacology & Chemical Biology, and University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, 2.32A Hillman Cancer Center Research Pavilion, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA
| | | |
Collapse
|
194
|
Wróbel AM, Gregoraszczuk EŁ. Differential effect of methyl-, butyl- and propylparaben and 17β-estradiol on selected cell cycle and apoptosis gene and protein expression in MCF-7 breast cancer cells and MCF-10A non-malignant cells. J Appl Toxicol 2014; 34:1041-50. [DOI: 10.1002/jat.2978] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 11/21/2013] [Accepted: 11/21/2013] [Indexed: 01/27/2023]
Affiliation(s)
- Anna Maria Wróbel
- Department of Physiology and Toxicology of Reproduction Institute of Zoology; Jagiellonian University in Kraków; Gronostajowa 9 30-387 Krakow Poland
| | - Ewa Łucja Gregoraszczuk
- Department of Physiology and Toxicology of Reproduction Institute of Zoology; Jagiellonian University in Kraków; Gronostajowa 9 30-387 Krakow Poland
| |
Collapse
|
195
|
Pinto G, Alhaiek AAM, Amadi S, Qattan AT, Crawford M, Radulovic M, Godovac-Zimmermann J. Systematic nucleo-cytoplasmic trafficking of proteins following exposure of MCF7 breast cancer cells to estradiol. J Proteome Res 2014; 13:1112-27. [PMID: 24422525 DOI: 10.1021/pr4012359] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We have used a proteomics subcellular spatial razor approach to look at changes in total protein abundance and in protein distribution between the nucleus and cytoplasm following exposure of MCF7 breast cancer cells to estradiol. The dominant response of MCF7 cells to estrogen stimulation involves dynamic changes in protein subcellular spatial distribution rather than changes in total protein abundance. Of the 3604 quantitatively monitored proteins, only about 2% show substantial changes in total abundance (>2-fold), whereas about 20% of the proteins show substantial changes in local abundance and/or redistribution of their subcellular location, with up to 16-fold changes in their local concentration in the nucleus or the cytoplasm. We propose that dynamic redistribution of the subcellular location of multiple proteins in response to stimuli is a fundamental characteristic of cells and suggest that perturbation of cellular spatial control may be an important feature of cancer.
Collapse
Affiliation(s)
- Gabriella Pinto
- Proteomics and Molecular Cell Dynamics, Center for Nephrology, Division of Medicine, School of Life and Medical Sciences, University College London , Royal Free Campus, Rowland Hill Street, London NW3 2PF, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
196
|
Xuan QJ, Wang JX, Nanding A, Wang ZP, Liu H, Lian X, Zhang QY. Tumor-associated macrophages are correlated with tamoxifen resistance in the postmenopausal breast cancer patients. Pathol Oncol Res 2014; 20:619-24. [PMID: 24414992 DOI: 10.1007/s12253-013-9740-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Accepted: 12/12/2013] [Indexed: 02/06/2023]
Abstract
Tumor-associated macrophages (TAMs) have been correlated with increased angiogenesis and poor prognosis in breast cancer. However, the precise role of TAMs in tamoxifen resistance remains unclear. We used immunohistochemical method to examine the expression of epidermal growth factor receptor (EGFR) and CD163+ macrophages in 100 breast cancer tissues. The clinical and biological features of 100 patients were estrogen receptor (ER)-positive and human epidermal growth factor receptor 2(Her-2)-negative tumors. The tamoxifen resistant tissues (n = 48) were the surgical excision samples from patients who developed recurrence or metastasis at the time of adjuvant tamoxifen treatment. The tamoxifen resistant tissues were contrast to tamoxifen sensitive tissues (n = 52). Positive staining for EGFR and CD163+ macrophages were observed in 21 samples (43.8 %) and in 26 samples (54.2 %) respectively in tamoxifen resistance group, which were higher than that of tamoxifen sensitive group (P = 0.001 and P = 0.000279 respectively). Significant positive correlations were found between the expression of EGFR and CD163+ macrophages (r = 0.567, P < 0.01). CD163+ macrophages were positively correlated with tumor size, lymph node metastasis and obesity. Obesity was also related to tamoxifen resistance (P < 0.05). The patients with higher density of CD163+ macrophages infiltration suffered from shorter time to develop recurrence or metastasis (P < 0.05). TAMs may be associated with tamoxifen resistance. Further studies are needed to investigate the potential mechanism between TAMs and tamoxifen resistance.
Collapse
Affiliation(s)
- Qi-jia Xuan
- Department of Medical Oncology, Tumor Hospital of Harbin Medical University, Haping Road 150 of Nangang District, Harbin, 150081, Heilongjiang, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
197
|
Mott NN, Pinceti E, Rao YS, Przybycien-Szymanska MM, Prins SA, Shults CL, Yang X, Glucksman MJ, Roberts JL, Pak TR. Age-dependent Effects of 17β-estradiol on the dynamics of estrogen receptor β (ERβ) protein-protein interactions in the ventral hippocampus. Mol Cell Proteomics 2014; 13:760-79. [PMID: 24390426 DOI: 10.1074/mcp.m113.031559] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Recent clinical evidence suggests that the neuroprotective and beneficial effects of hormone therapy may be limited by factors related to age and reproductive status. The patient's age and length of time without circulating ovarian hormones are likely to be key factors in the specific neurological outcomes of hormone therapy. However, the mechanisms underlying age-related changes in hormone efficacy have not been determined. We hypothesized that there are intrinsic changes in estrogen receptor β (ERβ) function that determine its ability to mediate the actions of 17β-estradiol (E2) in brain regions such as the ventral hippocampus. In this study, we identified and quantified a subset of ERβ protein interactions in the ventral hippocampus that were significantly altered by E2 replacement in young and aged animals, using two-dimensional differential gel electrophoresis coupled with liquid chromatography-electrospray ionization-tandem mass spectrometry. This study demonstrates quantitative changes in ERβ protein-protein interactions with E2 replacement that are dependent upon age in the ventral hippocampus and how these changes could alter processes such as transcriptional regulation. Thus, our data provide evidence that changes in ERβ protein interactions are a potential mechanism for age-related changes in E2 responsiveness in the brain after menopause.
Collapse
Affiliation(s)
- Natasha N Mott
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois 60153
| | | | | | | | | | | | | | | | | | | |
Collapse
|
198
|
Calaf GM, Balajee AS, Montalvo-Villagra MT, Leon M, Daniela NM, Alvarez RG, Roy D, Narayan G, Abarca-Quinones J. Vimentin and Notch as biomarkers for breast cancer progression. Oncol Lett 2014; 7:721-727. [PMID: 24527079 PMCID: PMC3919898 DOI: 10.3892/ol.2014.1781] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Accepted: 11/06/2013] [Indexed: 01/27/2023] Open
Abstract
Breast cancer, the most common spontaneous malignancy diagnosed in women, is a classical model of hormone dependency as it is associated with prolonged exposure to female hormones. Different cytoplasmic proteins are important in the transformation of a normal cell to an invasive tumor cell, and these include vimentin and Notch. To investigate the importance of these two genes and proteins in breast carcinogenesis, we used an in vitro breast cancer model system, in which an immortalized human breast epithelial cell line, MCF-10F, was malignantly transformed by exposure to low doses of high linear energy transfer α particle (150 keV/μm) radiation and subsequent growth in the presence or absence of 17β-estradiol. This model consisted of human breast epithelial cells in different stages of transformation: i) a parental cell line (MCF-10F), ii) an Estrogen cell line (MCF-l0F continuously grown with estradiol at 10−8), iii) a malignant and non-tumorigenic cell line (Alpha3), iv) a malignant and tumorigenic cell line (Alpha5) and v) a Tumor2 cell line derived from a xenograft of the Alpha5 cell line injected into nude mice. Vimentin and Notch showed greater expression in the Alpha5 and Tumor2 cell lines compared with that in the non-tumorigenic cell lines, MCF-10F, Estrogen and Alpha3. In the present study, positive staining for vimentin was found in 21% of cases. Vimentin and Notch protein expression was negative in noninvasive ductal carcinoma biopsies from breast cancer patients. However, positive cell expression was observed in invasive ductal carcinoma biopsies. These biomarkers can be considered important indicators of breast cancer progression and can be added to the diagnostic panel when overall survival is a primary end-point.
Collapse
Affiliation(s)
- Gloria M Calaf
- Institute for Advanced Research, Tarapacá University, Arica 8097877, Chile ; Center for Radiological Research, Columbia University Medical Center, New York, NY 10032, USA
| | - Adayabalam S Balajee
- Center for Radiological Research, Columbia University Medical Center, New York, NY 10032, USA
| | | | - Mariana Leon
- Departament of Health Science, Faculty of Science, Tarapacá University, Arica 8097877, Chile
| | - Navarrete M Daniela
- Department of Pathology, Dr. Gustavo Fricke Hospital of Viña del Mar, Valparaiso 2581907, Chile
| | - Raúl González Alvarez
- Department of Pathology, Dr. Gustavo Fricke Hospital of Viña del Mar, Valparaiso 2581907, Chile
| | - Debasish Roy
- Department of Natural Sciences, Hostos College of the City University of New York, Bronx, NY 10451, USA
| | - Gopeshwar Narayan
- Department of Human Molecular Genetics, Banaras Hindu University, Varanasi 221005, India
| | - Jorge Abarca-Quinones
- IREC, School of Medicine, IREC, St. Luc Hospital, University of Louvain, Brussels 1200, Belgium
| |
Collapse
|
199
|
Zou JX, Duan Z, Wang J, Sokolov A, Xu J, Chen CZ, Li JJ, Chen HW. Kinesin family deregulation coordinated by bromodomain protein ANCCA and histone methyltransferase MLL for breast cancer cell growth, survival, and tamoxifen resistance. Mol Cancer Res 2014; 12:539-49. [PMID: 24391143 DOI: 10.1158/1541-7786.mcr-13-0459] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
UNLABELLED Kinesins are a superfamily of motor proteins and often deregulated in different cancers. However, the mechanism of their deregulation has been poorly understood. Through examining kinesin gene family expression in estrogen receptor (ER)-positive breast cancer cells, we found that estrogen stimulation of cancer cell proliferation involves a concerted regulation of specific kinesins. Estrogen strongly induces expression of 19 kinesin genes such as Kif4A/4B, Kif5A/5B, Kif10, Kif11, Kif15, Kif18A/18B, Kif20A/20B, Kif21, Kif23, Kif24, Kif25, and KifC1, whereas suppresses the expression of seven others, including Kif1A, Kif1C, Kif7, and KifC3. Interestingly, the bromodomain protein ANCCA/ATAD2, previously shown to be an estrogen-induced chromatin regulator, plays a crucial role in the up- and downregulation of kinesins by estrogen. Its overexpression drives estrogen-independent upregulation of specific kinesins. Mechanistically, ANCCA (AAA nuclear coregulator cancer associated) mediates E2-dependent recruitment of E2F and MLL1 histone methyltransferase at kinesin gene promoters for gene activation-associated H3K4me3 methylation. Importantly, elevated levels of Kif4A, Kif15, Kif20A, and Kif23 correlate with that of ANCCA in the tumors and with poor relapse-free survival of patients with ER-positive breast cancer. Their knockdown strongly impeded proliferation and induced apoptosis of both tamoxifen-sensitive and resistant cancer cells. Together, the study reveals ANCCA as a key mediator of kinesin family deregulation in breast cancer and the crucial role of multiple kinesins in growth and survival of the tumor cells. IMPLICATIONS These findings support the development of novel inhibitors of cancer-associated kinesins and their regulator ANCCA for effective treatment of cancers including tamoxifen-resistant breast cancers.
Collapse
Affiliation(s)
- June X Zou
- UC Davis Cancer Center/Basic Sciences, University of California, Davis, UCDMC Research III, Room 1400B, 4645 2nd Avenue, Sacramento, CA 95817.
| | | | | | | | | | | | | | | |
Collapse
|
200
|
Pérez-Hernández AI, Catalán V, Gómez-Ambrosi J, Rodríguez A, Frühbeck G. Mechanisms linking excess adiposity and carcinogenesis promotion. Front Endocrinol (Lausanne) 2014; 5:65. [PMID: 24829560 PMCID: PMC4013474 DOI: 10.3389/fendo.2014.00065] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 04/15/2014] [Indexed: 12/17/2022] Open
Abstract
Obesity constitutes one of the most important metabolic diseases being associated to insulin resistance development and increased cardiovascular risk. Association between obesity and cancer has also been well established for several tumor types, such as breast cancer in post-menopausal women, colorectal, and prostate cancer. Cancer is the first death cause in developed countries and the second one in developing countries, with high incidence rates around the world. Furthermore, it has been estimated that 15-20% of all cancer deaths may be attributable to obesity. Tumor growth is regulated by interactions between tumor cells and their tissue microenvironment. In this sense, obesity may lead to cancer development through dysfunctional adipose tissue and altered signaling pathways. In this review, three main pathways relating obesity and cancer development are examined: (i) inflammatory changes leading to macrophage polarization and altered adipokine profile; (ii) insulin resistance development; and (iii) adipose tissue hypoxia. Since obesity and cancer present a high prevalence, the association between these conditions is of great public health significance and studies showing mechanisms by which obesity lead to cancer development and progression are needed to improve prevention and management of these diseases.
Collapse
Affiliation(s)
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
- *Correspondence: Gema Frühbeck, Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Avda. Pío XII 36, Pamplona 31008, Spain e-mail:
| |
Collapse
|