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










Publication year range
1.
Nat Commun ; 7: 10751, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26916619

ABSTRACT

Recent high-throughput studies revealed recurrent RUNX1 mutations in breast cancer, specifically in oestrogen receptor-positive (ER(+)) tumours. However, mechanisms underlying the implied RUNX1-mediated tumour suppression remain elusive. Here, by depleting mammary epithelial cells of RUNX1 in vivo and in vitro, we demonstrate combinatorial regulation of AXIN1 by RUNX1 and oestrogen. RUNX1 and ER occupy adjacent elements in AXIN1's second intron, and RUNX1 antagonizes oestrogen-mediated AXIN1 suppression. Accordingly, RNA-seq and immunohistochemical analyses demonstrate an ER-dependent correlation between RUNX1 and AXIN1 in tumour biopsies. RUNX1 loss in ER(+) mammary epithelial cells increases ß-catenin, deregulates mitosis and stimulates cell proliferation and expression of stem cell markers. However, it does not stimulate LEF/TCF, c-Myc or CCND1, and it does not accelerate G1/S cell cycle phase transition. Finally, RUNX1 loss-mediated deregulation of ß-catenin and mitosis is ameliorated by AXIN1 stabilization in vitro, highlighting AXIN1 as a potential target for the management of ER(+) breast cancer.


Subject(s)
Axin Protein/genetics , Breast Neoplasms/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Estrogens/metabolism , Gene Expression Regulation, Neoplastic , Receptors, Estrogen/genetics , beta Catenin/metabolism , Animals , Axin Protein/metabolism , Blotting, Western , Breast Neoplasms/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Cyclin D1 , Female , G1 Phase Cell Cycle Checkpoints , Gene Expression Profiling , Humans , Immunohistochemistry , MCF-7 Cells , Mice , Proto-Oncogene Proteins c-myc , Receptors, Estrogen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , TCF Transcription Factors
2.
Int J Cancer ; 137(4): 797-809, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25648303

ABSTRACT

The potential role of osteoblasts in bone and bone marrow (BM) metastases in neuroblastoma (NBL) remains unclear. In this study, we examined the effect of NBL cells on the osteoblastic differentiation of BM-derived mesenchymal stromal cells (BMMSC). We show that the presence of NBL cells enhanced the osteoblastic differentiation of BMMSC driven by bone morphogenetic protein (BMP)-4, in the absence of any effect on NBL cell proliferation. Expression profiles of BMMSC driven toward osteoblastic differentiation revealed an increase in vascular endothelial growth factor A (Vegfa) expression in the presence of NBL cells. We demonstrated that NBL cells increased BMMSC-derived VEGFA mRNA and protein and that this was enhanced by BMP-4. However, in similar conditions, neither the addition of an mVEGFA blocking antibody nor exogenous recombinant (r) mVEGFA affected osteoblastic differentiation. In contrast, siRNA- mediated knock-down of VEGFA in BMMSC prevented osteoblastic differentiation in BMP-4-treated cocultures, an effect that was not reversed in the presence of rmVEGFA. An analysis of murine bones injected with hNBL cells revealed an increase of mVEGFA producing cells near tumor cells concomitantly with an increase in Vegfa and Runx2 mRNA. This coincided with an increase in osteoclasts, in Rankl/Opg mRNA ratio and with the formation of osteolytic lesions. Thus NBL cells promote osteoblastogenesis in the BM by increasing VEGFA expression in BMMSC. Our study provides a new insight into the role of VEGFA in NBL metastases by pointing to the role of stroma-derived intracrine VEGFA in osteoblastogenesis.


Subject(s)
Cell Differentiation/genetics , Lymphocyte Activation/genetics , Mesenchymal Stem Cells/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Bone Morphogenetic Protein 4/administration & dosage , Cell Line , Cell Proliferation/genetics , Core Binding Factor Alpha 1 Subunit/biosynthesis , Gene Expression Regulation, Developmental , Humans , Mice , Neuroblastoma/metabolism , Osteoblasts/metabolism , Osteoprotegerin/biosynthesis , RNA, Messenger/biosynthesis , Vascular Endothelial Growth Factor A/genetics
3.
Mol Endocrinol ; 28(4): 546-53, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24606124

ABSTRACT

Excessive bone resorption is the cause of several metabolic bone diseases including osteoporosis. Thus, identifying factors that can inhibit osteoclast formation and/or activity may define new drug targets that can be used to develop novel therapies for these conditions. Emerging evidence demonstrates that the master regulator of hematopoiesis, Runx1, is expressed in preosteoclasts and may influence skeletal health. To examine the potential role of Runx1 in osteoclast formation and function, we deleted its expression in myeloid osteoclast precursors by crossing Runx1 floxed mice (Runx1(F/F)) with CD11b-Cre transgenic mice. Mice lacking Runx1 in preosteoclasts (CD11b-Cre;Runx1(F/F)) exhibited significant loss of femoral trabecular and cortical bone mass compared with that in Cre-negative mice. In addition, serum levels of collagen type 1 cross-linked C-telopeptide, a biomarker of osteoclast-mediated bone resorption, were significantly elevated in CD11b-Cre;Runx1(F/F) mice compared with those in Runx1(F/F) mice. Tartrate-resistant acid phosphatase-positive osteoclasts that differentiated from bone marrow cells of CD11b-Cre;Runx1(F/F) mice in vitro were larger, were found in greater numbers, and had increased bone resorbing activity than similarly cultured cells from Runx1(F/F) mice. CD11b-Cre;Runx1(F/F) bone marrow cells that were differentiated into osteoclasts in vitro also had elevated mRNA levels of osteoclast-related genes including vacuolar ATPase D2, cathepsin K, matrix metalloproteinase 9, calcitonin receptor, osteoclast-associated receptor, nuclear factor of activated T cells cytoplasmic 1, and cFos. These data indicate that Runx1 expression in preosteoclasts negatively regulates osteoclast formation and activity and contributes to overall bone mass.


Subject(s)
Cell Differentiation , Core Binding Factor Alpha 2 Subunit/metabolism , Osteoclasts/pathology , Animals , Bone Marrow Cells/metabolism , Bone Resorption/metabolism , Bone Resorption/pathology , Bone and Bones/metabolism , Bone and Bones/pathology , CD11b Antigen/metabolism , Gene Deletion , Integrases/metabolism , Mice , Organ Size , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/metabolism , Osteogenesis
4.
Cancer Res ; 74(10): 2857-68, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24648349

ABSTRACT

Changes to androgen signaling during prostate carcinogenesis are associated with both inhibition of cellular differentiation and promotion of malignant phenotypes. The androgen receptor (AR)-binding transcription factor RUNX2 has been linked to prostate cancer progression but the underlying mechanisms have not been fully defined. In this study, we investigated the genome-wide influence of RUNX2 on androgen-induced gene expression and AR DNA binding in prostate cancer cells. RUNX2 inhibited the androgen response partly by promoting the dissociation of AR from its target genes such as the tumor suppressor NKX3-1. However, AR activity persists in the presence of RUNX2 at other AR target genes, some of which are cooperatively stimulated by androgen and RUNX2 signaling. These genes are associated with putative enhancers co-occupied by AR and RUNX2. One such gene, the invasion-promoting Snail family transcription factor SNAI2, was co-activated by AR and RUNX2. Indeed, these two transcription factors together, but neither alone stimulated prostate cancer cell invasiveness, which could be abolished by SNAI2 silencing. Furthermore, an immunohistochemical analysis of SNAI2 in archived primary prostate cancer specimens revealed a correlation with the RUNX2 histoscore, and simultaneous strong staining for SNAI2, RUNX2, and AR (but not any pair alone) was associated with disease recurrence. Overall, our findings suggest cooperation between AR and RUNX in the stimulation of oncogenes such as SNAI2, which might be targeted for individualized prostate cancer therapy.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Transcription Factors/genetics , Animals , Biopsy , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Core Binding Factor Alpha 1 Subunit/genetics , Dihydrotestosterone/pharmacology , Doxycycline/pharmacology , Gene Expression/drug effects , Humans , Male , Neoplasm Invasiveness , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Snail Family Transcription Factors , Transcription Factors/biosynthesis
5.
J Cell Biochem ; 115(1): 27-33, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23943595

ABSTRACT

The efficacy of glucocorticoids (GCs) in treating a wide range of autoimmune and inflammatory conditions is blemished by severe side effects, including osteoporosis. The chief mechanism leading to GC-induced osteoporosis is inhibition of bone formation, but the role of RUNX2, a master regulator of osteoblast differentiation and bone formation, has not been well studied. We assessed effects of the synthetic GC dexamethasone (dex) on transcription of RUNX2-stimulated genes during the differentiation of mesenchymal pluripotent cells into osteoblasts. Dex inhibited a RUNX2 reporter gene and attenuated locus-dependently RUNX2-driven expression of several endogenous target genes. The anti-RUNX2 activity of dex was not attributable to decreased RUNX2 expression, but rather to physical interaction between RUNX2 and the GC receptor (GR), demonstrated by co-immunoprecipitation assays and co-immunofluorescence imaging. Investigation of the RUNX2/GR interaction may lead to the development of bone-sparing GC treatment modalities for the management of autoimmune and inflammatory diseases.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Glucocorticoids/pharmacology , Mesenchymal Stem Cells/cytology , Osteoblasts/drug effects , Alkaline Phosphatase/antagonists & inhibitors , Alkaline Phosphatase/metabolism , Animals , Cell Differentiation/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Dexamethasone/pharmacology , Mesenchymal Stem Cells/drug effects , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Receptors, Glucocorticoid/metabolism
6.
PLoS One ; 8(6): e62361, 2014.
Article in English | MEDLINE | ID: mdl-23755096

ABSTRACT

Prolactin-induced Protein (PIP), an aspartyl protease unessential for normal mammalian cell function, is required for the proliferation and invasion of some breast cancer (BCa) cell types. Because PIP expression is particularly high in the Luminal A BCa subtype, we investigated the roles of PIP in the related T47D BCa cell line. Nucleic acid and antibody arrays were employed to screen effects of PIP silencing on global gene expression and activation of receptor tyrosine kinases (RTKs), respectively. Expression of PIP-stimulated genes, as defined in the T47D cell culture model, was well correlated with the expression of PIP itself across a cohort of 557 mRNA profiles of diverse BCa tumors, and bioinformatics analysis revealed cJUN and cMYC as major nodes in the PIP-dependent gene network. Among 71 RTKs tested, PIP silencing resulted in decreased phosphorylation of focal adhesion kinase (FAK), ephrin B3 (EphB3), FYN, and hemopoietic cell kinase (HCK). Ablation of PIP also abrogated serum-induced activation of the downstream serine/threonine kinases AKT, ERK1/2, and JNK1. Consistent with these results, PIP-depleted cells exhibited defects in adhesion to fibronectin, cytoskeletal stress fiber assembly and protein secretion. In addition, PIP silencing abrogated the mitogenic response of T47D BCa cells to estradiol (E2). The dependence of BCa cell proliferation was unrelated, however, to estrogen signaling because: 1) PIP silencing did not affect the transcriptional response of estrogen target genes to hormone treatment, and 2) PIP was required for the proliferation of tamoxifen-resistant BCa cells. Pharmacological inhibition of PIP may therefore serve the bases for both augmentation of existing therapies for hormone-dependent tumors and the development of novel therapeutic approaches for hormone-resistant BCa.


Subject(s)
Breast Neoplasms/classification , Breast Neoplasms/pathology , Carrier Proteins/metabolism , Estrogens/pharmacology , Glycoproteins/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Carrier Proteins/genetics , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Glycoproteins/genetics , HEK293 Cells , Humans , Membrane Transport Proteins , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Stress Fibers/drug effects , Stress Fibers/metabolism , Tamoxifen/pharmacology , Transcriptome/drug effects , Transcriptome/genetics
7.
J Cell Biochem ; 113(7): 2406-14, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22389001

ABSTRACT

Runx2, best known for its role in regulating osteoblast-specific gene expression, also plays an increasingly recognized role in prostate and breast cancer metastasis. Using the C4-2B/Rx2(dox) prostate cancer cell line that conditionally expressed Runx2 in response to doxycycline treatment, we identified and characterized G9a, a histone methyltransferase, as a novel regulator for Runx2 activity. G9a function was locus-dependent. Whereas depletion of G9a reduced expression of many Runx2 target genes, including MMP9, CSF2, SDF1, and CST7, expression of others, such as MMP13 and PIP, was enhanced. Physical association between G9a and Runx2 was indicated by co-immunoprecipitation, GST-pulldown, immunofluorescence, and fluorescence recovery after photobleaching (FRAP) assays. Since G9a makes repressive histone methylation marks and is primarily known as a corepressor, we further investigated the mechanism by which G9a functioned as a positive regulator for Runx2 target genes. Transient reporter assays indicated that the histone methyltransferase activity of G9a was not required for transcriptional activation by Runx2. Chromatin immunoprecipitation assays for Runx2 and G9a showed that G9a was recruited to endogenous Runx2 binding sites. We conclude that a subset of cancer-related Runx2 target genes require recruitment of G9a for their expression, but do not depend on its histone methyltransferase activity.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Transcription, Genetic , Animals , COS Cells , Cell Line, Tumor , Chemokine CXCL12/biosynthesis , Chlorocebus aethiops , Cystatins/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Humans , Male , Matrix Metalloproteinase 13/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Promoter Regions, Genetic , Prostatic Neoplasms , Transcriptional Activation
8.
J Cell Physiol ; 227(4): 1776-85, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21732355

ABSTRACT

Transcellular bicarbonate transport is suspected to be an important pathway used by ameloblasts to regulate extracellular pH and support crystal growth during enamel maturation. Proteins that play a role in amelogenesis include members of the ABC transporters (SLC gene family and CFTR). A number of carbonic anhydrases (CAs) have also been identified. The defined functions of these genes are likely interlinked during enamel mineralization. The purpose of this study is to quantify relative mRNA levels of individual SLC, Cftr, and CAs in enamel cells obtained from secretory and maturation stages on rat incisors. We also present novel data on the enamel phenotypes for two animal models, a mutant porcine (CFTR-ΔF508) and the NBCe1-null mouse. Our data show that two SLCs (AE2 and NBCe1), Cftr, and Car2, Car3, Car6, and Car12 are all significantly up-regulated at the onset of the maturation stage of amelogenesis when compared to the secretory stage. The remaining SLCs and CA gene transcripts showed negligible expression or no significant change in expression from secretory to maturation stages. The enamel of CFTR-ΔF508 adult pigs was hypomineralized and showed abnormal crystal growth. NBCe1-null mice enamel was structurally defective and had a marked decrease in mineral content relative to wild-type. These data demonstrate the importance of many non-matrix proteins to amelogenesis and that the expression levels of multiple genes regulating extracellular pH are modulated during enamel maturation in response to an increased need for pH buffering during hydroxyapatite crystal growth.


Subject(s)
Dental Enamel/growth & development , Dental Enamel/metabolism , Amelogenesis/genetics , Amelogenesis/physiology , Animals , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Antiporters/genetics , Antiporters/metabolism , Base Sequence , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , DNA Primers/genetics , Dental Enamel/abnormalities , Hydrogen-Ion Concentration , Ion Transport , Male , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Mutant Proteins/genetics , Mutant Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , SLC4A Proteins , Sodium-Bicarbonate Symporters/deficiency , Sodium-Bicarbonate Symporters/genetics , Sodium-Bicarbonate Symporters/metabolism , Sodium-Calcium Exchanger/genetics , Sus scrofa
9.
J Cell Physiol ; 227(5): 2276-82, 2012 May.
Article in English | MEDLINE | ID: mdl-21809344

ABSTRACT

Prolactin-Induced Protein (PIP) is a small polypeptide expressed by breast and prostate cancer (BCa, PCa) cells. However, both the regulation of PIP expression and its function in cancer cells are poorly understood. Using BCa and PCa cells, we found that Runx2, a pro-metastatic transcription factor, functionally interacts with the Androgen Receptor (AR) to regulate PIP expression. Runx2 expression in C4-2B PCa cells synergized with AR to promote PIP expression, whereas its knockdown in T47D BCa cells abrogated basal as well as hormone stimulated PIP expression. Chromatin immunoprecipitation (ChIP) assays showed that Runx2 and AR co-occupied an enhancer element located ∼11 kb upstream of the PIP open reading frame, and that Runx2 facilitated AR recruitment to the enhancer. PIP knockdown in T47D cells compromised DHT-stimulated expression of multiple AR target genes including PSA, FKBP5, FASN, and SGK1. The inhibition of AR activity due to loss of PIP was attributable at least in part to abrogation of its nuclear translocation. PIP knockdown also suppressed T47D cell proliferation driven by either serum growth factors or dihydrotestosterone (DHT). Our data suggest that Runx2 controls a positive feedback loop between androgen signaling and PIP, and pharmacological inhibition of PIP may be useful to treat PIP positive tumors.


Subject(s)
Androgens/pharmacology , Carrier Proteins/pharmacology , Cell Proliferation/drug effects , Core Binding Factor Alpha 1 Subunit/metabolism , Feedback, Physiological/physiology , Glycoproteins/pharmacology , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Membrane Transport Proteins , Microarray Analysis/methods , Receptors, Androgen/metabolism
10.
Nucleic Acids Res ; 40(8): 3538-47, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22187159

ABSTRACT

Runx2 is a metastatic transcription factor (TF) increasingly expressed during prostate cancer (PCa) progression. Using PCa cells conditionally expressing Runx2, we previously identified Runx2-regulated genes with known roles in epithelial-mesenchymal transition, invasiveness, angiogenesis, extracellular matrix proteolysis and osteolysis. To map Runx2-occupied regions (R2ORs) in PCa cells, we first analyzed regions predicted to bind Runx2 based on the expression data, and found that recruitment to sites upstream of the KLK2 and CSF2 genes was cyclical over time. Genome-wide ChIP-seq analysis at a time of maximum occupancy at these sites revealed 1603 high-confidence R2ORs, enriched with cognate motifs for RUNX, GATA and ETS TFs. The R2ORs were distributed with little regard to annotated transcription start sites (TSSs), mainly in introns and intergenic regions. Runx2-upregulated genes, however, displayed enrichment for R2ORs within 40 kb of their TSSs. The main annotated functions enriched in 98 Runx2-upregulated genes with nearby R2ORs were related to invasiveness and membrane trafficking/secretion. Indeed, using SDS-PAGE, mass spectrometry and western analyses, we show that Runx2 enhances secretion of several proteins, including fatty acid synthase and metastasis-associated laminins. Thus, combined analysis of Runx2's transcriptome and genomic occupancy in PCa cells lead to defining its novel role in regulating protein secretion.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Prostatic Neoplasms/genetics , Binding Sites , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/physiology , DNA/chemistry , DNA/metabolism , DNA, Intergenic/metabolism , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Introns , Male , Nucleotide Motifs , Prostatic Neoplasms/metabolism , Proteins/genetics , Proteins/metabolism , Up-Regulation
11.
Clin Cancer Res ; 18(3): 901-11, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22147940

ABSTRACT

PURPOSE: To assess the clinical significance of the interaction between estrogen and Runx2 signaling, previously shown in vitro. EXPERIMENTAL DESIGN: MCF7/Rx2(dox) breast cancer cells were treated with estradiol and/or doxycycline to induce Runx2, and global gene expression was profiled to define genes regulated by estradiol, Runx2, or both. Anchorage-independent growth was assessed by soft-agar colony formation assays. Expression of gene sets defined using the MCF7/Rx2(dox) system was analyzed in pre- and on-treatment biopsies from hormone receptor-positive patients undergoing neoadjuvant letrozole treatment in two independent studies, and short-term changes in gene expression were correlated with tumor size reduction or Ki67 index at surgery. RESULTS: Reflecting its oncogenic property, estradiol strongly promoted soft-agar colony formation, whereas Runx2 blocked this process suggesting tumor suppressor property. Transcriptome analysis of MCF7/Rx2(dox) cells treated with estradiol and/or doxycycline showed reciprocal attenuation of Runx2 and estrogen signaling. Correspondingly in breast cancer tumors, expression of estradiol- and Runx2-regulated genes was inversely correlated, and letrozole increased expression of Runx2-stimulated genes, as defined in the MCF7/Rx2(dox) model. Of particular interest was a gene set upregulated by estradiol and downregulated by Runx2 in vitro; its short-term response to letrozole treatment associated with tumor size reduction and Ki67 index at surgery better than other estradiol-regulated gene sets. CONCLUSION: This work provides clinical evidence for the importance of antagonism between Runx2 and E2 signaling in breast cancer. Likely sensing the tension between them, letrozole responsiveness of a genomic node, positively regulated by estradiol and negatively regulated by Runx2 in vitro, best correlated with the clinical efficacy of letrozole treatment.


Subject(s)
Breast Neoplasms/genetics , Cell Proliferation , Core Binding Factor Alpha 1 Subunit/genetics , Drug Resistance, Neoplasm/genetics , Estradiol/genetics , Gene Expression Regulation, Neoplastic/genetics , Antineoplastic Agents/therapeutic use , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Clinical Trials, Phase II as Topic , Core Binding Factor Alpha 1 Subunit/metabolism , Estradiol/metabolism , Female , Fluorescent Antibody Technique , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Letrozole , Nitriles/therapeutic use , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Triazoles/therapeutic use
12.
Breast Cancer Res ; 13(6): R127, 2011.
Article in English | MEDLINE | ID: mdl-22151997

ABSTRACT

INTRODUCTION: In contrast to its role in breast cancer (BCa) initiation, estrogen signaling has a protective effect in later stages, where estrogen receptor (ER)α loss associates with aggressive metastatic disease. We asked whether the beneficial effect of estrogen signaling in late-stage BCa is attributable to the recently reported estrogen-mediated antagonism of the pro-metastatic transcription factor Runx2. METHODS: MCF7/Rx2dox breast cancer cells were engineered with a lentivirus expressing Runx2 in response to doxycycline (dox). Cells treated with dox and/or estradiol (E2) were subjected to genome-wide expression profiling, RT-qPCR analysis of specific genes, and Matrigel™ invasion assays. Knockdown of genes of interest was performed using lentiviruses expressing appropriate shRNAs, either constitutively or in response to dox. Gene expression in BCa tumors was investigated using a cohort of 557 patients compiled from publicly available datasets. Association of gene expression with clinical metastasis was assessed by dichotomizing patients into those expressing genes of interest at either high or low levels, and comparing the respective Kaplan-Meier curves of metastasis-free survival. RESULTS: Runx2 induced epithelial-mesenchymal transition (EMT) evidenced by acquisition of a fibroblastic morphology, decreased expression of E-cadherin, increased expression of vimentin and invasiveness. Runx2 stimulated SNAI2 expression in a WNT- and transforming growth factor (TGF)ß-dependent manner, and knockdown of SNAI2 abrogated the pro-metastatic activities of Runx2. E2 antagonized the pro-metastatic activities of Runx2, including SNAI2 upregulation. In primary BCa tumors, Runx2 activity, SNAI2 expression, and metastasis were positively correlated, and SNAI2 expression was negatively correlated with ERα. However, the negative correlation between SNAI2 and ERα in bone-seeking BCa cells was weaker than the respective negative correlation in tumors seeking lung. Furthermore, the absence of ERα in primary tumors was associated with lung- and brain- but not with bone metastasis, and tumor biopsies from bone metastatic sites displayed the unusual combination of high Runx2/SNAI2 and high ERα expression. CONCLUSIONS: E2 antagonizes Runx2-induced EMT and invasiveness of BCa cells, partly through attenuating expression of SNAI2, a Runx2 target required for mediating its pro-metastatic property. That ERα loss promotes non-osseous metastasis by unleashing Runx2/SNAI2 is supported by the negative correlation observed in corresponding tumors. Unknown mechanisms in bone-seeking BCa allow high Runx2/SNAI2 expression despite high ERα level.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Core Binding Factor Alpha 1 Subunit/metabolism , Estrogens/metabolism , Signal Transduction , Transcription Factors/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Estrogens/pharmacology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , Snail Family Transcription Factors , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism
13.
Mol Cancer ; 9: 258, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20863401

ABSTRACT

BACKGROUND: Prostate cancer (PCa) cells preferentially metastasize to bone at least in part by acquiring osteomimetic properties. Runx2, an osteoblast master transcription factor, is aberrantly expressed in PCa cells, and promotes their metastatic phenotype. The transcriptional programs regulated by Runx2 have been extensively studied during osteoblastogenesis, where it activates or represses target genes in a context-dependent manner. However, little is known about the gene regulatory networks influenced by Runx2 in PCa cells. We therefore investigated genome wide mRNA expression changes in PCa cells in response to Runx2. RESULTS: We engineered a C4-2B PCa sub-line called C4-2B/Rx2 dox, in which Doxycycline (Dox) treatment stimulates Runx2 expression from very low to levels observed in other PCa cells. Transcriptome profiling using whole genome expression array followed by in silico analysis indicated that Runx2 upregulated a multitude of genes with prominent cancer associated functions. They included secreted factors (CSF2, SDF-1), proteolytic enzymes (MMP9, CST7), cytoskeleton modulators (SDC2, Twinfilin, SH3PXD2A), intracellular signaling molecules (DUSP1, SPHK1, RASD1) and transcription factors (Sox9, SNAI2, SMAD3) functioning in epithelium to mesenchyme transition (EMT), tissue invasion, as well as homing and attachment to bone. Consistent with the gene expression data, induction of Runx2 in C4-2B cells enhanced their invasiveness. It also promoted cellular quiescence by blocking the G1/S phase transition during cell cycle progression. Furthermore, the cell cycle block was reversed as Runx2 levels declined after Dox withdrawal. CONCLUSIONS: The effects of Runx2 in C4-2B/Rx2 dox cells, as well as similar observations made by employing LNCaP, 22RV1 and PC3 cells, highlight multiple mechanisms by which Runx2 promotes the metastatic phenotype of PCa cells, including tissue invasion, homing to bone and induction of high bone turnover. Runx2 is therefore an attractive target for the development of novel diagnostic, prognostic and therapeutic approaches to PCa management. Targeting Runx2 may prove more effective than focusing on its individual downstream genes and pathways.


Subject(s)
Bone Neoplasms/secondary , Core Binding Factor Alpha 1 Subunit/metabolism , Prostatic Neoplasms/complications , Prostatic Neoplasms/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Apoptosis/genetics , Apoptosis/physiology , Biomarkers, Tumor/genetics , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation , Chemokine CXCL12/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Cystatins/genetics , Dual Specificity Phosphatase 1/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Male , Matrix Metalloproteinase 9/genetics , Microfilament Proteins/genetics , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/genetics , Protein-Tyrosine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Syndecan-2/genetics
14.
Blood ; 116(19): 3964-71, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20716776

ABSTRACT

Krox20/EGR2, one of the 4 early growth response genes, is a highly conserved transcription factor implicated in hindbrain development, peripheral nerve myelination, tumor suppression, and monocyte/macrophage cell fate determination. Here, we established a novel role for Krox20 in postnatal skeletal metabolism. Microcomputed tomographic analysis of 4- and 8-week-old mice revealed a low bone mass phenotype (LBM) in both the distal femur and the vertebra of Krox20(+/-) mice. This was attributable to accelerated bone resorption as demonstrated in vivo by increased osteoclast number and serum C-terminal telopeptides, a marker for collagen degradation. Krox20 haploinsufficiency did not reduce bone formation in vivo, nor did it compromise osteoblast differentiation in vitro. In contrast, growth and differentiation were significantly stimulated in preosteoclast cultures derived from Krox20(+/-) splenocytes, suggesting that the LBM is attributable to Krox20 haploinsufficiency in the monocytic lineage. Furthermore, Krox20 silencing in preosteoclasts increased cFms expression and response to macrophage colony-stimulating factor, leading to a cell-autonomous stimulation of cell-cycle progression. Our data indicate that the antimitogenic role of Krox20 in preosteoclasts is the predominant mechanism underlying the LBM phenotype of Krox20-deficient mice. Stimulation of Krox20 expression in preosteoclasts may present a viable therapeutic strategy for high-turnover osteoporosis.


Subject(s)
Bone and Bones/metabolism , Early Growth Response Protein 2/deficiency , Monocytes/cytology , Monocytes/metabolism , Osteoporosis/etiology , Animals , Base Sequence , Bone Resorption/etiology , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Cycle , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Proliferation , DNA Primers/genetics , Disease Models, Animal , Early Growth Response Protein 2/genetics , Female , Haploinsufficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , RNA, Small Interfering/genetics , Signal Transduction
15.
J Cell Physiol ; 224(2): 305-10, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20432458

ABSTRACT

Recent reports reveal increasing complexity of mechanisms underlying the bone sparing effects of sex steroids. This review focuses on mechanisms by which sex steroids attenuate endocortical and trabecular adult bone turnover, perhaps their most important property as bone mass regulators. Clearly, estrogen withdrawal increases osteoclast number and bone resorption; however, important open questions are the extent to which osteoblasts and their precursors are involved, and the relative contributions of the RANK/RANKL/OPG system, Fas ligand and Runx2. In addition to reviewing these aspects of estrogen action, we also discuss proskeletal effects of androgens on the adult male skeleton, including aromatization to estrogens and male-specific mechanisms. Detailed understanding of skeletal site- and gender-dependent mechanisms by which sex steroids protect the adult skeleton will provide the foundation for improved risk assessment, prevention and management of osteoporosis.


Subject(s)
Bone Remodeling/physiology , Gonadal Steroid Hormones/metabolism , Steroids/metabolism , Adult , Animals , Humans , Models, Animal , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism
16.
J Biol Chem ; 284(31): 20848-57, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19491106

ABSTRACT

In plants, a family of more than 20 heat stress transcription factors (Hsf) controls the expression of heat stress (hs) genes. There is increasing evidence for the functional diversification between individual members of the Hsf family fulfilling distinct roles in response to various environmental stress conditions and developmental signals. In response to hs, accumulation of both heat stress proteins (Hsp) and Hsfs is induced. In tomato, the physical interaction between the constitutively expressed HsfA1 and the hs-inducible HsfA2 results in synergistic transcriptional activation (superactivation) of hs gene expression. Here, we show that the interaction is strikingly specific and not observed with other class A Hsfs. Hetero-oligomerization of the two-component Hsfs is preferred to homo-oligomerization, and each Hsf in the HsfA1/HsfA2 hetero-oligomeric complex has its characteristic contribution to its function as superactivator. Distinct regions of the oligomerization domain are responsible for specific homo- and hetero-oligomeric interactions leading to the formation of hexameric complexes. The results are summarized in a model of assembly and function of HsfA1/A2 superactivator complexes in hs gene regulation.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Plant , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Heat-Shock Response/genetics , Multiprotein Complexes/metabolism , Plant Proteins/chemistry , Plant Proteins/metabolism , Solanum lycopersicum/genetics , Stress, Physiological/genetics , Transcription Factors/chemistry , Transcription Factors/metabolism , Amino Acid Motifs , Amino Acid Sequence , Chromatography, Gel , Cross-Linking Reagents/metabolism , DNA-Binding Proteins/genetics , Heat Shock Transcription Factors , Heat-Shock Proteins/genetics , Models, Biological , Molecular Sequence Data , Plant Proteins/genetics , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , Transcription Factors/genetics
17.
Mol Endocrinol ; 23(8): 1203-14, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19389811

ABSTRACT

Runx2 and androgen receptor (AR) are master transcription factors with pivotal roles in bone metabolism and prostate cancer (PCa). We dissected AR-mediated repression of Runx2 in dihydrotestosterone (DHT)-treated osteoblastic and PCa cells using reporter assays and endogenous Runx2 target genes. Repression required DHT, but not AR's transactivation function, and was associated with nuclear colocalization of the two proteins. Runx2 and AR coimmunoprecipitated and interacted directly in glutathione-S-transferase pull-down assays. Interaction was ionic in nature. Intact AR DNA-binding domain (DBD) was necessary and sufficient for both interaction with Runx2 and its repression. Runx2 sequences required for interaction were the C-terminal 132 amino acid residues together with the Runt DBD. Runx2 DNA binding was abrogated by endogenous AR in chromatin immunoprecipitation assays and by recombinant AR-DBD in gel shift assays. Furthermore, AR caused increased nuclear mobility of Runx2 as indicated by faster fluorescence recovery after photobleaching. Thus, AR binds Runx2 and abrogates its binding to DNA and possibly to other nuclear components. Clinical relevance of our results was suggested by an inverse correlation between expression of AR-responsive prostate-specific antigen and osteocalcin genes in PCa biopsies. Given the tumor suppressor properties of Runx2, its repression by AR may constitute a mechanism of hormone carcinogenesis. Attenuation of Runx2 by AR in osteoblasts may play a role in skeletal metabolism: the bone-sparing effect of androgens is attributable, in part, to keeping Runx2 activity in check and preventing high-turnover bone disease such as seen after castration and in transgenic mice overexpressing Runx2 in osteoblasts.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Osteoblasts/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , 3T3 Cells , Animals , Bone and Bones/metabolism , COS Cells , Chlorocebus aethiops , DNA/metabolism , Humans , Male , Mice , Mice, Transgenic , Transcription Factors/metabolism
18.
J Cell Biochem ; 105(4): 965-70, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18821584

ABSTRACT

The mRNAs encoding Runx2, a master osteoblast transcription factor, and its target gene Osteocalcin (OC), are commonly used as markers of osteoblast differentiation. We found that while OC mRNA levels do indeed increase during development of the osteoblast phenotype in MC3T3-E1 cultures, Runx2 mRNA levels surprisingly decrease. Neither translational control of Runx2 (based on Western analysis) nor regulation of its DNA-binding ability (assessed by electrophoretic mobility shift assay) could explain the unexpected opposite patterns of Runx2 and OC expression. Instead, a series of chromatin immunoprecipitation (ChIP) assays during osteoblast differentiation revealed that early on, when Runx2 protein amount and DNA-binding activity are maximal, it is practically absent from the OC promoter. At later stages, Runx2 is recruited to the OC promoter while Runx2 mRNA, protein, and in vitro DNA binding progressively decrease. We also followed Runx2 occupancy at a novel genomic target discovered by ChIP-Chip analysis of cells in which the OC promoter is maximally occupied. The results revealed that Runx2 is recruited to this locus and to the OC promoter with a remarkably similar temporal pattern. These observations highlight a mechanism that restrains Runx2-mediated transcriptional control by confining its access to genomic targets to a narrow window of time. The need for such stringent control is consistent with the severe consequences of Runx2 over-expression in vivo.


Subject(s)
Cell Differentiation , Core Binding Factor Alpha 1 Subunit/genetics , Gene Expression Regulation, Developmental , Osteoblasts/cytology , Osteocalcin/genetics , Promoter Regions, Genetic , Animals , Cell Line , Core Binding Factor Alpha 1 Subunit/metabolism , Mice , Protein Transport , RNA, Messenger/analysis , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Endocrinology ; 149(12): 5984-95, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18755791

ABSTRACT

The transcription factors Runx2 and estrogen receptor-alpha (ERalpha) are involved in numerous normal and disease processes, including postmenopausal osteoporosis and breast cancer. Using indirect immunofluorescence microscopy and pull-down techniques, we found them to colocalize and form complexes in a ligand-dependent manner. Estradiol-bound ERalpha strongly interacted with Runx2 directly through its DNA-binding domain and only indirectly through its N-terminal and ligand-binding domains. Runx2's amino acids 417-514, encompassing activation domain 3 and the nuclear matrix targeting sequence, were sufficient for interaction with ERalpha's DNA-binding domain. As a consequence of the interaction, Runx2's transcriptional activation activity was strongly repressed, as shown by reporter assays in COS7 cells, breast cancer cells, and late-stage MC3T3-E1 osteoblast cultures. Metaanalysis of gene expression in 779 breast cancer biopsies indicated negative correlation between the expression of ERalpha and Runx2 target genes. Selective ER modulators (SERM) induced ERalpha-Runx2 interactions but led to various functional outcomes. The regulation of Runx2 by ERalpha may play key roles in osteoblast and breast epithelial cell growth and differentiation; hence, modulation of Runx2 by native and synthetic ERalpha ligands offers new avenues in selective ER modulator evaluation and development.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Estrogen Receptor alpha/metabolism , Osteoblasts/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , COS Cells , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Core Binding Factor Alpha 1 Subunit/genetics , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Gene Expression/drug effects , Immunoprecipitation , Mice , Oligonucleotide Array Sequence Analysis , Osteoblasts/cytology , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , Protein Binding/drug effects , Reverse Transcriptase Polymerase Chain Reaction
20.
J Biol Chem ; 282(6): 3605-13, 2007 Feb 09.
Article in English | MEDLINE | ID: mdl-17150959

ABSTRACT

Unlike other eukaryotes, plants possess a complex family of heat stress transcription factors (Hsfs) with usually more than 20 members. Among them, Hsfs A4 and A5 form a group distinguished from other Hsfs by structural features of their oligomerization domains and by a number of conserved signature sequences. We show that A4 Hsfs are potent activators of heat stress gene expression, whereas A5 Hsfs act as specific repressors of HsfA4 activity. The oligomerization domain of HsfA5 alone is necessary and sufficient to exert this effect. Due to the high specificity of the oligomerization domains, other class A Hsfs are not affected. Pull-down assay and yeast two-hybrid interaction tests demonstrate that the tendency to form HsfA4/A5 heterooligomers is stronger than the formation of homooligomers. The specificity of interaction between Hsfs A4 and A5 was confirmed by bimolecular fluorescence complementation experiments. The major role of the representatives of the HsfA4/A5 group, which are not involved in the conventional heat stress response, may reside in cell type-specific functions connected with the control of cell death triggered by pathogen infection and/or reactive oxygen species.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/physiology , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/physiology , Plant Proteins/antagonists & inhibitors , Plant Proteins/physiology , Repressor Proteins/physiology , Solanum lycopersicum/physiology , Transcription Factors/antagonists & inhibitors , Transcription Factors/physiology , DNA-Binding Proteins/metabolism , Heat Shock Transcription Factors , Heat-Shock Proteins/metabolism , Plant Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...