Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Anticancer Res ; 40(2): 767-777, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32014919

ABSTRACT

BACKGROUND/AIM: Chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) plays an important role in cancer. We examined the effect of COUP-TFII overexpression on the proliferation and invasion of the human colorectal cancer SNU-C4 cells. MATERIALS AND METHODS: SNU-C4 cells were stably transfected with COUP-TFII expression plasmid to overexpress COUP-TFII (COUP-TFII-SNU-C4 cells). Cell proliferation, colony-forming ability and transwell invasion assays were performed. To elucidate the underlying molecular mechanism of COUP-TFII action, western blot analysis, p53 shRNA transfection, and Myr-Akt transfection were performed. RESULTS: Cell proliferation and colony-forming ability were significantly inhibited in COUP-TFII-SNU-C4 cells. Western blot analyses demonstrated that while the expression of p53 and PTEN was increased, the p-Akt levels were decreased in COUP-TFII-SNU-C4 cells. Knockdown of p53 partially restored the cell proliferation, but did not reverse the inhibition of invasion. Constitutive activation of Akt via Myr-Akt transfection reversed the inhibited cell proliferation and invasion by COUP-TFII. CONCLUSION: p53 is required for the inhibition of cell proliferation, and decreased phosphorylation of Akt may mediate the inhibition of cell proliferation and invasion by COUP-TFII.


Subject(s)
COUP Transcription Factor II/biosynthesis , Colorectal Neoplasms/metabolism , PTEN Phosphohydrolase/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Protein p53/biosynthesis , COUP Transcription Factor II/metabolism , Cell Proliferation/physiology , Colorectal Neoplasms/pathology , Humans , PTEN Phosphohydrolase/metabolism , Phosphorylation , Transfection , Tumor Suppressor Protein p53/metabolism
2.
Int J Cancer ; 139(7): 1574-85, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27193872

ABSTRACT

COUP-TFII belongs to the nuclear receptor family, which is highly expressed in many kinds of tumors. Previous studies have shown that COUP-TFII can promote tumor progression through regulating tumor angiogenesis and cell proliferation and migration of certain cancer cells. However, the function of COUP-TFII in renal cell carcinoma (RCC) is not clear. Here, we showed that clinical RCC tumor tissues showed much higher COUP-TFII expression level than adjacent normal tissues. When COUP-TFII was knocked down in RCC 769-P and 786-O cells by siRNA or shRNA-expressing lentivirus, the cell proliferation was markedly inhibited, and apoptosis increased. Moreover, the tumor growth of COUP-TFII knockdown 769-P and 786-O xenografts in nude mice was also obviously inhibited. Using qRT-PCR and Western blot, we showed that the expression of the tumor suppressor gene BRCA1 was upregulated in COUP-TFII knockdown cells. Simultaneously knockdown of BRCA1 and COUP-TFII partially rescued the inhibited cell proliferation and increased apoptosis in COUP-TFII single knockdown cells. These results indicate that COUP-TFII may play an oncogenic role in RCC, and COUP-TFII may promote tumor progression through inhibiting BRCA1.


Subject(s)
BRCA1 Protein/genetics , COUP Transcription Factor II/genetics , Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Animals , Apoptosis/immunology , BRCA1 Protein/biosynthesis , COUP Transcription Factor II/biosynthesis , COUP Transcription Factor II/deficiency , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Knockdown Techniques , Genes, BRCA1 , Heterografts , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Up-Regulation
3.
Mol Endocrinol ; 29(9): 1362-74, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26241389

ABSTRACT

Steroid hormone-regulated differentiation of uterine stromal cells, known as decidualization, is essential for embryo implantation. The role of the estrogen receptor-α (ESR1) during this differentiation process is unclear. Development of conditional Esr1-null mice showed that deletion of this gene in both epithelial and stromal compartments of the uterus leads to a complete blockade of decidualization, indicating a critical role of ESR1 during this process. To further elucidate the cell type-specific function of ESR1 in the uterus, we created WE(d/d) mice in which Esr1 is ablated in uterine luminal and glandular epithelia but is retained in the stroma. Uteri of WE(d/d) mice failed to undergo decidualization, indicating that epithelial ESR1 contributes to stromal differentiation via a paracrine mechanism. We noted markedly reduced production of the leukemia inhibitory factor (LIF) in WE(d/d) uteri. Supplementation with LIF restored decidualization in WE(d/d) mice. Our study indicated that LIF acts synergistically with progesterone to induce the expression of Indian hedgehog (IHH) in uterine epithelium and its receptor patched homolog 1 in the stroma. IHH then induces the expression of chicken ovalbumin upstream promoter-transcription factor II, a transcription factor that promotes stromal differentiation. To address the mechanism by which LIF induces IHH expression, we used mice lacking uterine epithelial signal transducer and activator of transcription 3, a well-known mediator of LIF signaling. Our study revealed that LIF-mediated induction of IHH occurs without the activation of epithelial signal transducer and activator of transcription 3 but uses an alternate pathway involving the activation of the ERK1/2 kinase. Collectively our results provide unique insights into the paracrine mechanisms by which ESR1 directs epithelial-stromal dialogue during pregnancy establishment.


Subject(s)
Decidua/growth & development , Embryo Implantation/physiology , Estrogen Receptor alpha/metabolism , Leukemia Inhibitory Factor/metabolism , Paracrine Communication/physiology , Animals , COUP Transcription Factor II/biosynthesis , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Decidua/cytology , Decidua/metabolism , Enzyme Activation/genetics , Estrogen Receptor alpha/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Hedgehog Proteins/biosynthesis , Leukemia Inhibitory Factor/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucous Membrane/cytology , Mucous Membrane/metabolism , Patched Receptors , Patched-1 Receptor , Pregnancy , Progesterone/metabolism , Receptors, Cell Surface/biosynthesis , STAT3 Transcription Factor/genetics , Signal Transduction/genetics
4.
Neuromolecular Med ; 17(4): 353-63, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26208914

ABSTRACT

Mutations in the gene encoding the transcriptional modulator methyl-CpG binding protein 2 (MeCP2) are responsible for the neurodevelopmental disorder Rett syndrome which is one of the most frequent sources of intellectual disability in women. Recent studies showed that loss of Mecp2 in astrocytes contributes to Rett-like symptoms and restoration of Mecp2 can rescue some of these defects. The goal of this work is to compare gene expression profiles of wild-type and mutant astrocytes from Mecp2(308/y) mice (B6.129S-MeCP2/J) by using Affymetrix mouse 2.0 microarrays. Results were confirmed by quantitative real-time RT-PCR and by Western blot analysis. Gene set enrichment analysis utilizing Ingenuity Pathways was employed to identify pathways disrupted by Mecp2 deficiency. A total of 2152 genes were statistically differentially expressed between wild-type and mutated samples, including 1784 coding transcripts. However, only 257 showed fold changes >1.2. We confirmed our data by replicative studies in independent primary cultures of cortical astrocytes from Mecp2-deficient mice. Interestingly, two genes known to encode secreted proteins, chromogranin B and lipocalin-2, showed significant dysregulation. These proteins secreted from Mecp2-deficient glia may exert negative non-cell autonomous effects on neuronal properties, including dendritic morphology. Moreover, transcriptional profiling revealed altered Nr2f2 expression which may explain down- and upregulation of several target genes in astrocytes such as Ccl2, Lcn2 and Chgb. Unraveling Nr2f2 involvement in Mecp2-deficient astrocytes could pave the way for a better understanding of Rett syndrome pathophysiology and offers new therapeutic perspectives.


Subject(s)
Astrocytes/metabolism , Methyl-CpG-Binding Protein 2/deficiency , Nerve Tissue Proteins/genetics , Rett Syndrome/genetics , Transcriptome , Acute-Phase Proteins/metabolism , Animals , COUP Transcription Factor II/biosynthesis , COUP Transcription Factor II/genetics , Cells, Cultured , Cerebral Cortex/pathology , Chromogranin B/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Genome-Wide Association Study , Lipocalin-2 , Lipocalins/metabolism , Male , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Inbred Strains , NF-kappa B/metabolism , Nerve Tissue Proteins/biosynthesis , Oligonucleotide Array Sequence Analysis , Oncogene Proteins/metabolism , RNA Interference , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Rett Syndrome/pathology
5.
Int J Cancer ; 134(7): 1648-58, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24122412

ABSTRACT

Despite the accumulating knowledge of alterations in pancreatic cancer molecular pathways, no substantial improvements in the clinical prognosis have been made and this malignancy continues to be a leading cause of cancer death in the Western World. The orphan nuclear receptor COUP-TFII is a regulator of a wide range of biological processes and it may exert a pro-oncogenic role in cancer cells; interestingly, indirect evidences suggest that the receptor could be involved in pancreatic cancer. The aim of this study was to evaluate the expression of COUP-TFII in human pancreatic tumors and to unveil its role in the regulation of pancreatic tumor growth. We evaluated COUP-TFII expression by immunohistochemistry on primary samples. We analyzed the effect of the nuclear receptor silencing in human pancreatic cancer cells by means of shRNA expressing cell lines. We finally confirmed the in vitro results by in vivo experiments on nude mice. COUP-TFII is expressed in 69% of tested primary samples and correlates with the N1 and M1 status and clinical stage; Kaplan-Meier and Cox regression analysis show that it may be an independent prognostic factor of worst outcome. In vitro silencing of COUP-TFII reduces the cell growth and invasiveness and it strongly inhibits angiogenesis, an effect mediated by the regulation of VEGF-C. In nude mice, COUP-TFII silencing reduces tumor growth by 40%. Our results suggest that COUP-TFII might be an important regulator of the behavior of pancreatic adenocarcinoma, thus representing a possible new target for pancreatic cancer therapy.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , COUP Transcription Factor II/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Adenocarcinoma/blood supply , Adenocarcinoma/metabolism , Aged , Animals , COUP Transcription Factor II/biosynthesis , Cell Growth Processes/physiology , Cell Line, Tumor , Disease Progression , Female , Humans , Male , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/metabolism , Prognosis , Vascular Endothelial Growth Factor C/biosynthesis , Vascular Endothelial Growth Factor C/genetics , Pancreatic Neoplasms
6.
Proc Natl Acad Sci U S A ; 110(11): 4333-8, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23440210

ABSTRACT

The nuclear receptor Chicken Ovalbumin Upstream Promoter-Transcription Factor II (COUP-TFII) is an important coordinator of glucose homeostasis through its function in different organs such as the endocrine pancreas, adipose tissue, skeletal muscle, and liver. Recently we have demonstrated that COUP-TFII expression in the hypothalamus is restricted to a subpopulation of neurons expressing the steroidogenic factor 1 transcription factor, known to play a crucial role in glucose homeostasis. To understand the functional significance of COUP-TFII expression in the steroidogenic factor 1 neurons, we generated hypothalamic ventromedial nucleus-specific COUP-TFII KO mice using the cyclization recombination/locus of X-overP1 technology. The heterozygous mutant mice display insulin hypersensitivity and a leaner phenotype associated with increased energy expenditure and similar food intake. These mutant mice also present a defective counterregulation to hypoglycemia with altered glucagon secretion. Moreover, the mutant mice are more likely to develop hypoglycemia-associated autonomic failure in response to recurrent hypoglycemic or glucopenic events. Therefore, COUP-TFII expression levels in the ventromedial nucleus are keys in the ability to resist the onset of hypoglycemia-associated autonomic failure.


Subject(s)
COUP Transcription Factor II/biosynthesis , Glucose/metabolism , Hypoglycemia/metabolism , Nerve Tissue Proteins/biosynthesis , Neurons/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Autonomic Nervous System Diseases/etiology , Autonomic Nervous System Diseases/genetics , Autonomic Nervous System Diseases/metabolism , Autonomic Nervous System Diseases/pathology , COUP Transcription Factor II/genetics , Chickens , Glucose/genetics , Heterozygote , Hypoglycemia/complications , Hypoglycemia/genetics , Hypoglycemia/pathology , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/pathology , Organ Specificity/genetics , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Ventromedial Hypothalamic Nucleus/pathology
7.
J Comp Neurol ; 521(2): 479-97, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-22791192

ABSTRACT

Neocortical GABAergic interneurons in rodents originate from subpallial progenitor zones. The majority of mouse neocortical interneurons are derived from the medial and caudal ganglionic eminences (MGE and CGE, respectively) and the preoptic area (POA). It is controversial whether the lateral ganglionic eminence (LGE) also generates neocortical interneurons. Previously it was shown that the transcription factor COUP-TFII is expressed in the CGE; here we show that COUP-TFII is also expressed in the dorsal MGE, dorsal LGE (dMGE and dLGE, respectively), and POA. In the adult neocortex, COUP-TFII+/somatostatin (SOM)+ interneurons are mainly located in layer V. Using a genetic fate-mapping approach (Shh-Cre and Nkx2.1-Cre), we demonstrate that the POA and ventral MGE do not give rise to COUP-TFII+ neocortical interneurons, suggesting that the dMGE is the source of COUP-TFII+/SOM+ neocortical interneurons. We also observed a migratory stream of COUP-TFII+/Sp8+ cells emanating from the dLGE and CGE to the neocortex mainly through the subventricular zone at later embryonic stages. Slice culture experiments in which dLGE progenitors were labeled with BrdU provided additional evidence that the dLGE generates neocortical interneurons. While earlier-born dMGE-derived COUP-TFII+ interneurons occupy cortical layer V, later-born dLGE- and CGE-derived COUP-TFII+ ones preferentially occupy superficial cortical layers. A similar laminar distribution was observed following neonatal transplantation of embryonic day (E)14.5 dMGE and E15.5 dLGE. These results provide novel information about interneuron fate and position from spatially and temporally distinct origins in the ganglionic eminences.


Subject(s)
COUP Transcription Factor II/biosynthesis , COUP Transcription Factor II/genetics , Caudate Nucleus/physiology , Interneurons/physiology , Neocortex/physiology , Animals , Antibodies , Antimetabolites , Bromodeoxyuridine , Caudate Nucleus/embryology , Cell Transplantation , Female , Gene Expression Regulation, Developmental , Immunohistochemistry , Mice , Mice, Inbred Strains , Microscopy, Confocal , Neocortex/cytology , Neocortex/embryology , Organ Culture Techniques , Pregnancy , Rats , Rats, Wistar , Stem Cells/physiology
8.
Differentiation ; 84(4): 330-43, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22906706

ABSTRACT

The heterogeneous nature of stem cells is an important issue in both research and therapeutic use in terms of directing cell lineage differentiation pathways, as well as self-renewal properties. Using flow cytometry we have identified two distinct subpopulations by size, large and small, within cultures of human embryonic stem (hES) cell lines. These two cell populations respond differentially to retinoic acid (RA) differentiation and several endocrine disruptor compounds (EDC). The large cell population responds to retinoic acid differentiation with greater than a 50% reduction in cell number and loss of Oct-4 expression, whereas the number of the small cell population does not change and Oct-4 protein expression is maintained. In addition, four estrogenic compounds altered SSEA-3 expression differentially between the two cell subpopulations changing their ratios relative to each other. Both populations express stem cell markers Oct-4, Nanog, Tra-1-60, Tra-1-80 and SSEA-4, but express low levels of differentiation markers common to the three germ layers. Cloning studies indicate that both populations can revive the parental population. Furthermore, whole genome microarray identified approximately 400 genes with significantly different expression between the two populations (p<0.01). We propose the differential response to RA in these populations is due to differential gene expression of Notch signaling members, CoupTF1 and CoupTF2, chromatin remodeling and histone modifying genes that render the small population resistant to RA differentiation. The findings that hES cells exist as heterogeneous populations with distinct responses to differentiation signals and environmental stimuli will be relevant for their use for drug discovery and disease therapy.


Subject(s)
Cell Differentiation/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Endocrine Disruptors/pharmacology , Tretinoin/pharmacology , Antigens, Differentiation/metabolism , Antigens, Surface/biosynthesis , Antigens, Tumor-Associated, Carbohydrate/biosynthesis , Apigenin/pharmacology , COUP Transcription Factor II/biosynthesis , Cell Cycle/drug effects , Cell Line , Chlordecone/pharmacology , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Homeodomain Proteins/biosynthesis , Humans , Kaempferols/pharmacology , Nanog Homeobox Protein , Octamer Transcription Factor-3/biosynthesis , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Proteoglycans/biosynthesis , Signal Transduction/drug effects , Stage-Specific Embryonic Antigens/biosynthesis , Tamoxifen/pharmacology
9.
Basic Res Cardiol ; 106(6): 1269-81, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22057716

ABSTRACT

Despite the apparent appropriateness of left ventricular (LV) remodeling following myocardial infarction (MI), it poses an independent risk factor for development of heart failure. There is a paucity of studies into the molecular mechanisms of LV remodeling in large animal species. We took an unbiased molecular approach to identify candidate transcription factors (TFs) mediating the genetic reprogramming involved in post-MI LV remodeling in swine. Left ventricular tissue was collected from remote, non-infarcted myocardium, 3 weeks after MI-induction or sham-surgery. Microarray analysis identified 285 upregulated and 278 downregulated genes (FDR < 0.05). Of these differentially expressed genes, the promoter regions of the human homologs were searched for common TF binding sites (TFBS). Eighteen TFBS were overrepresented >two-fold (p < 0.01) in upregulated and 13 in downregulated genes. Left ventricular nuclear protein extracts were assayed for DNA-binding activity by protein/DNA array. Out of 345 DNA probes, 30 showed signal intensity changes >two-fold. Five TFs were identified in both TFBS and protein/DNA array analyses, which showed matching changes for COUP-TFII and glucocorticoid receptor (GR) only. Treatment of swine with the GR antagonist mifepristone after MI reduced the post-MI increase in LV mass, but LV dilation remained unaffected. Thus, using an unbiased approach to study post-MI LV remodeling in a physiologically relevant large animal model, we identified COUP-TFII and GR as potential key mediators of post-MI remodeling.


Subject(s)
Gene Expression Profiling , Myocardial Infarction/genetics , Ventricular Remodeling/genetics , Animals , COUP Transcription Factor II/biosynthesis , COUP Transcription Factor II/genetics , Disease Models, Animal , Echocardiography , Female , Genomics , Male , Myocardial Infarction/physiopathology , Oligonucleotide Array Sequence Analysis , Receptors, Glucocorticoid/biosynthesis , Receptors, Glucocorticoid/genetics , Swine , Transcription Factors
10.
Oncol Rep ; 21(1): 101-6, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19082449

ABSTRACT

Chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) plays an essential role in angiogenesis and development. A previous study showed that the expression of COUP-TFII enhanced invasiveness of human lung carcinoma cells. However, no published data are available concerning the biological and clinical significance of COUP-TFII expression in colorectal cancer. Thus, our objective was to explore the expression of COUP-TFII in colorectal cancer as well as its association with clinicopathological features, and to evaluate the role of COUP-TFII as a prognostic indicator in colorectal cancer. We investigated the presence of COUP-TFII in human colorectal cancer tissues and adjacent normal tissues from 95 primary colorectal cancer patients by immunohistochemistry. The correlation between the expression of COUP-TFII and clinicopathologic features was investigated. The 3-year disease-free survival (DFS) and overall survival (OS) of patients with tumors expressing different levels of COUP-TFII were evaluated by the Kaplan-Meier method. No significant correlation was found between COUP-TFII expression and age at surgery, gender, histopathologic differentiation, vessel invasion, carcinoembryonic antigen (CEA), or nodal involvement. However, survival analysis showed that the COUP-TFII-positive group had a significantly better OS compared to COUP-TFII-negative group (80.4% vs. 57.7%, P=0.0491). Based on our results, COUP-TFII may represent a biomarker for good prognosis in colorectal cancer.


Subject(s)
Biomarkers, Tumor/analysis , COUP Transcription Factor II/biosynthesis , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease-Free Survival , Female , Gene Expression , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis
11.
J Neurosci ; 28(50): 13582-91, 2008 Dec 10.
Article in English | MEDLINE | ID: mdl-19074032

ABSTRACT

While the cortical interneurons derived from the medial ganglionic eminence (MGE) migrate rather diffusely into the cortex, interneurons that migrate out from the caudal ganglionic eminence (CGE) mainly move caudally into the caudal cerebral cortex and the hippocampus in the form of the caudal migratory stream (CMS) (Yozu et al., 2005). Although transplantation experiments at embryonic day 13.5 had revealed that the migrating cells in these two populations are already intrinsically different in regard to their ability to respond to the CGE environment (Yozu et al., 2005), it is not known how the CGE cells are specified and how their migratory behavior is determined. In this study we showed that, although CGE and lateral ganglionic eminence (LGE) express almost the same marker molecules, LGE cells do not migrate caudally when transplanted into the CGE, suggesting that LGE cells are intrinsically different from CGE cells. We therefore compared the transcriptomes of the CGE, MGE, and LGE, and the results showed that COUP-TFII was expressed preferentially in the CGE as well as in the migrating interneurons in the CMS. Transplantation experiments revealed that COUP-TFII is sufficient to change the direction of MGE cell migration to caudal when transplanted into the CGE environment, and knockdown of COUP-TFII inhibited the caudal migration of the CGE cells. These results suggest that COUP-TFII is both required and sufficient for the CGE-cell-specific migratory behavior in the caudal direction. Thus, a locally expressed transcription factor determines the migratory direction of the cortical interneurons in a region-specific manner.


Subject(s)
Body Patterning/physiology , COUP Transcription Factor II/biosynthesis , Cell Movement/physiology , Cerebral Cortex/embryology , Interneurons/metabolism , Animals , Brain Tissue Transplantation , Cerebral Cortex/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred ICR
12.
Cancer Res ; 66(20): 10188-98, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17047084

ABSTRACT

Tamoxifen (TAM) is successfully used for the treatment and prevention of breast cancer. However, many patients that are initially TAM responsive develop tumors that are antiestrogen/TAM resistant (TAM-R). The mechanism behind TAM resistance in estrogen receptor alpha (ERalpha)-positive tumors is not understood. The orphan nuclear receptor chicken ovalbumin upstream promoter transcription factor (COUP-TF)-I interacts directly with 4-hydroxytamoxifen (4-OHT)- and estradiol (E(2))-occupied ERalpha, corepressors NCoR and SMRT, and inhibit E(2)-induced gene transcription in breast cancer cells. Here we tested the hypothesis that reduced COUP-TFI and COUP-TFII correlate with TAM resistance. We report for the first time that COUP-TFII, but not COUP-TFI, is reduced in three antiestrogen/TAM-R cell lines derived from TAM-sensitive (TAM-S) MCF-7 human breast cancer cells and in MDA-MB-231 cells compared with MCF-7. ERalpha and ERbeta protein expression was not different between TAM-S and TAM-R cells, but progesterone receptor (PR) was decreased in TAM-R cells. Further, E(2) increased COUP-TFII transcription in MCF-7, but not TAM-R, cells. Importantly, reexpression of COUP-TFII in TAM-S cells to levels comparable to those in MCF-7 was shown to increase 4-OHT-mediated growth inhibition and increased apoptosis. Conversely, knockdown of COUP-TFII in TAM-S MCF-7 cells blocked growth inhibitory activity and increased 4-OHT agonist activity. 4-OHT increased COUP-TFII-ERalpha interaction approximately 2-fold in MCF-7 cells. COUP-TFII expression in TAM-R cells also inhibited 4-OHT-induced endogenous PR and pS2 mRNA expression. These data indicate that reduced COUP-TFII expression correlates with acquired TAM resistance in human breast cancer cell lines and that COUP-TFII plays a role in regulating the growth inhibitory activity of TAM in breast cancer cells.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/metabolism , COUP Transcription Factor II/biosynthesis , Tamoxifen/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , COUP Transcription Factor II/genetics , COUP Transcription Factor II/metabolism , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Chickens , Drug Resistance, Neoplasm , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/biosynthesis , Fulvestrant , Humans , Receptors, Progesterone/biosynthesis , Receptors, Progesterone/genetics , Tamoxifen/analogs & derivatives , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...