Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Sci Rep ; 9(1): 11802, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31413335

ABSTRACT

Proper wound healing is dynamic in order to maintain the corneal integrity and transparency. Impaired or delayed corneal epithelial wound healing is one of the most frequently observed ocular defect and difficult to treat. Cyclin dependen kinase (cdk), a known cell cycle regulator, required for proper proliferating and migration of cell. We therefore investigated the role of cell cycle regulator cdk10, member of cdk family and its functional association with transcriptional factor (ETS2) at active phase of corneal epithelial cell migration. Our data showed that cdk10 was associated with ETS2, while its expression was upregulated at the active phase (18 hours) of cell migration and gradually decrease as the wound was completely closed. Topical treatment with anti-cdk10 and ETS2 antibodies delayed the wound closure time at higest concentration (10 µg/ml) compared to control. Further, our results also showed increased mRNA expression of cdk10 and ETS2 at active phase of migration at approximately 2 fold. Collectively, our data reveals that cdk10 and ETS2 efficiently involved during corneal wound healing. Further studies are warranted to better understand the mechanism and safety of topical cdk10 and ETS2 proteins in corneal epithelial wound-healing and its potential role for human disease treatment.


Subject(s)
Corneal Injuries/pathology , Cyclin-Dependent Kinases/physiology , Epithelium, Corneal/pathology , Proto-Oncogene Protein c-ets-2/physiology , Wound Healing , Corneal Injuries/metabolism , Cyclin-Dependent Kinases/metabolism , Epithelium, Corneal/enzymology , Epithelium, Corneal/metabolism , Humans , In Vitro Techniques , Models, Biological , Proto-Oncogene Protein c-ets-2/metabolism
2.
Cancer Cell ; 31(1): 110-126, 2017 01 09.
Article in English | MEDLINE | ID: mdl-27989801

ABSTRACT

Tumor-associated endothelial cells (TECs) regulate tumor cell aggressiveness. However, the core mechanism by which TECs confer stem cell-like activity to indolent tumors is unknown. Here, we used in vivo murine and human tumor models to identify the tumor-suppressive checkpoint role of TEC-expressed insulin growth factor (IGF) binding protein-7 (IGFBP7/angiomodulin). During tumorigenesis, IGFBP7 blocks IGF1 and inhibits expansion and aggresiveness of tumor stem-like cells (TSCs) expressing IGF1 receptor (IGF1R). However, chemotherapy triggers TECs to suppress IGFBP7, and this stimulates IGF1R+ TSCs to express FGF4, inducing a feedforward FGFR1-ETS2 angiocrine cascade that obviates TEC IGFBP7. Thus, loss of IGFBP7 and upregulation of IGF1 activates the FGF4-FGFR1-ETS2 pathway in TECs and converts naive tumor cells to chemoresistant TSCs, thereby facilitating their invasiveness and progression.


Subject(s)
Insulin-Like Growth Factor Binding Proteins/physiology , Neoplastic Stem Cells/drug effects , Animals , Cell Transformation, Neoplastic , Drug Resistance, Neoplasm , Endothelial Cells/physiology , Fibroblast Growth Factor 4/physiology , Humans , Insulin-Like Growth Factor I/physiology , Mice , Proto-Oncogene Protein c-ets-2/physiology , Receptor, IGF Type 1/physiology
3.
Dev Biol ; 397(1): 77-88, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25446535

ABSTRACT

The ETS superfamily transcription factors Elf5 and Ets2 have both been implicated in the maintenance of the extraembryonic ectoderm (ExE) of the mouse embryo. While homozygous mutants of either gene result in various degrees of ExE tissue loss, heterozygotes are without phenotype. We show here that compound heterozygous mutants exhibit a phenotype intermediate to that of the more severe Elf5-/- and the milder Ets2-/- mutants. Functional redundancy is shown via commonalities in expression patterns, in target gene expression, and by partial rescue of Elf5-/- mutants through overexpressing Ets2 in an Elf5-like fashion. A model is presented suggesting the functional division of the ExE region into a proximal and distal domain based on gene expression patterns and the proximal to distal increasing sensitivity to threshold levels of combined Elf5 and Ets2 activity.


Subject(s)
DNA-Binding Proteins/physiology , Ectoderm/physiology , Gene Expression Regulation, Developmental , Proto-Oncogene Protein c-ets-2/physiology , Transcription Factors/physiology , Alleles , Animals , Animals, Genetically Modified , Cattle , Cell Differentiation , Fibroblasts/metabolism , Gene Expression Profiling , Heterozygote , Mice , Mutation , Oligonucleotide Array Sequence Analysis , Phenotype , RNA, Small Interfering/metabolism , Time Factors
4.
Curr Opin Hematol ; 19(3): 199-205, 2012 May.
Article in English | MEDLINE | ID: mdl-22406820

ABSTRACT

PURPOSE OF REVIEW: Vasculogenesis and hematopoiesis are essential for development. Recently, the ETS domain transcription factor Etv2 has been identified as an essential regulator of vasculogenesis and hematopoiesis. Here, we review the recent studies that have established the critical role of Etv2 in the specification of mesoderm to blood and endothelial cells. RECENT FINDINGS: Loss and gain-of-function studies have demonstrated the conserved role of Etv2 in endothelial and hematopoietic development. Recent studies have placed Etv2 at or near the top of the hierarchy in specification of these lineages and have begun to dissect the upstream regulators and downstream effectors of Etv2 function using multiple model organisms and experimental systems. SUMMARY: Etv2 is essential for the specification of endothelial and hematopoietic lineages. Understanding the mechanisms through which Etv2 specifies endothelial and blood cells by defining upstream transcriptional regulators and cofactors will lead to greater insight into vasculogenesis and hematopoiesis, and may help to identify therapeutic targets to treat vascular disorders or to promote or inhibit vessel growth.


Subject(s)
Endothelial Cells/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Neovascularization, Physiologic/physiology , Proto-Oncogene Protein c-ets-2/physiology , Cell Differentiation/physiology , Humans
5.
Carcinogenesis ; 33(4): 760-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22298639

ABSTRACT

E26 transformation-specific sequence (ETS)-2 is a transcriptional modulator located on chromosome 21, alterations in its expression have been implicated with a reduced incidence of solid tumors in Down syndrome patients. MicroRNAs (miRNAs) are thought to participate in diverse biological functions; however, the regulation of miRNAs is not well characterized. Recently, we reported that miR-196b is highly expressed in gastric cancers. Herein, we demonstrate that miR-196b expression was significantly repressed by ETS2 during gastric cancer oncogenesis. We demonstrate that knockdown of endogenous ETS2 expression increases miR-196b expression. A genomic region between -751 and -824 bp upstream of the miR-196b transcriptional start site was found to be critical for the repression activity. This putative regulatory promoter region contains three potential ETS2-binding motifs. Mutations within the ETS2 binding sites blocked the repression activity of ETS2. Furthermore, knockdown of ETS2 or overexpression of miR-196b significantly induced migration and invasion in gastric cancer cells. In addition, alterations in ETS2 and miR-196b expression in gastric cancer cell lines affected the expression of epithelial-mesenchymal transition-related genes. The levels of vimentin, matrix metalloproteinase (MMP)-2 and MMP9 were drastically induced, but levels of E-cadherin were decreased in shETS2- or miR-196b-transfected cells. Our data indicate that ETS2 plays a key role in controlling the expression of miR-196b, and miR-196b may mediate the tumor suppressor effects of ETS2. We demonstrated that miR-196b was transcriptionally regulated by ETS2 and there was an inverse expression profile between miR-196b and ETS2 in clinical samples. This finding could be beneficial for the development of effective cancer diagnostic and alternative therapeutic strategies.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Proto-Oncogene Protein c-ets-2/physiology , Stomach Neoplasms/genetics , Transcription, Genetic , Base Sequence , Cell Line, Tumor , Chromatin Immunoprecipitation , Humans , Molecular Sequence Data , Promoter Regions, Genetic , RNA Interference , Sequence Homology, Nucleic Acid , Stomach Neoplasms/pathology
6.
J Biol Chem ; 287(15): 11833-41, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22354960

ABSTRACT

Effector T cells and T cells from patients with systemic lupus erythematosus (SLE) express increased levels of the spleen tyrosine kinase (Syk). Syk binds to the T cell receptor (TCR)-CD3 complex and transduces the TCR-mediated signal in the cell more efficiently than the canonical CD3ζ chain. The reasons for the increased expression of Syk are unclear. In the present study, we found that Syk is regulated by the transcription factor c-Jun in cooperation with Ets2. c-Jun and Ets2 bound to the SYK promoter in close proximity and increased the promoter activity in a specific manner. Disruption of c-Jun and Ets2 expression by siRNA resulted in decreased expression of Syk. Overexpression of c-Jun but not Ets2 resulted in increase in Syk protein. c-Jun and Ets2 co-immunoprecipitated and had an additive effect on Syk expression. c-Jun-driven SYK promoter activation showed a similar pattern in B cells; however, as expected, basal promoter activity was much higher in B cells as compared with T cells. Overexpression of c-Jun led to increase in intracytoplasmic calcium flux following TCR stimulation. Moreover, we found that SLE T cells had increased levels of c-Jun at baseline and phosphorylated c-Jun upon activation. Finally, disruption of c-Jun and Ets2 in SLE T cells resulted in a decrease in calcium flux upon TCR stimulation. In conclusion, c-Jun in cooperation with Ets2 increases the expression of Syk and contributes to Syk-mediated heightened calcium responses in SLE T cells.


Subject(s)
Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/physiology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Protein c-ets-2/physiology , T-Lymphocytes/enzymology , Adult , B-Lymphocytes/enzymology , B-Lymphocytes/metabolism , Base Sequence , Binding Sites , Calcium Signaling , Cells, Cultured , Down-Regulation , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Lupus Erythematosus, Systemic/enzymology , Lupus Erythematosus, Systemic/pathology , Male , Middle Aged , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Protein c-ets-2/metabolism , Syk Kinase , T-Lymphocytes/metabolism , Transcription, Genetic
7.
FEBS Lett ; 585(15): 2537-44, 2011 Aug 04.
Article in English | MEDLINE | ID: mdl-21763315

ABSTRACT

Human cancers utilise telomerase to maintain telomeres and prohibit cell senescence. Human telomerase reverse transcriptase (hTERT), an essential component of this complex, is regulated at the level of gene transcription. Using SILAC-proteomic analysis and molecular studies, we identified the AAA+ ATPase, RuvBl2 as a transcriptional regulator of hTERT and established that this regulation is through cooperation with Ets-2. In colon cancer patients, nuclear expression of RuvBl2 associated with nuclear expression of hTERT, pEts2 and advanced nodal disease (P<0.01, P=0.05 and P=0.03 respectively, n=170). These data firmly implicate RuvBl2 in Ets2 mediated regulation of hTERT in colon cancer which has functional and clinical consequences.


Subject(s)
Carrier Proteins/physiology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA Helicases/physiology , Gene Expression Regulation, Neoplastic , Proto-Oncogene Protein c-ets-2/physiology , Telomerase/genetics , ATPases Associated with Diverse Cellular Activities , Colonic Neoplasms/chemistry , Disease Progression , Humans , Proteomics , Transcription, Genetic
8.
Stem Cells ; 29(3): 430-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21425406

ABSTRACT

Ets2 has both tumor repressive and supportive functions for different types of cancer. We have investigated the role of Ets2 within intestinal epithelial cells in postnatal mouse colon development and tumorigenesis. Conditional inactivation of Ets2 within intestinal epithelial cells results in over representation of Ets2-deficient colon crypts within young and adult animals. This preferential representation is associated with an increased number of proliferative cells within the stem cell region and an increased rate of crypt fission in young mice that result in larger patches of Ets2-deficient crypts. These effects are consistent with a selective advantage of Ets2-deficient intestinal stem cells in colonizing colonic crypts and driving crypt fission. Ets2-deficient colon crypts have an increased mucosal thickness, an increased number of goblet cells, and an increased density. Mice with Ets2-deficient intestinal cells develop more colon tumors in response to treatment with azoxymethane and dextran sulfate sodium. The selective population of colon crypts, the altered differentiation state and increased sensitivity to carcinogen-induced tumors all indicate that Ets2 deficiency alters colon stem cell number or behavior. Ets2-dependent, epithelial cell-autonomous repression of intestinal tumors may contribute to protection from colon cancer of persons with increased dosage of chromosome 21.


Subject(s)
Adenoma/genetics , Adult Stem Cells/pathology , Cell Transformation, Neoplastic/genetics , Colon/cytology , Colonic Neoplasms/genetics , Proto-Oncogene Protein c-ets-2/physiology , Adenoma/pathology , Adult Stem Cells/metabolism , Animals , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Colon/metabolism , Colon/pathology , Colonic Neoplasms/pathology , Down-Regulation/genetics , Down-Regulation/physiology , Female , Genetic Predisposition to Disease , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity/genetics , Proto-Oncogene Protein c-ets-2/genetics , Proto-Oncogene Protein c-ets-2/metabolism
9.
Dev Genes Evol ; 220(3-4): 107-15, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20680330

ABSTRACT

Convergent evolution of echinoderm pluteus larva was examined from the standpoint of functional evolution of a transcription factor Ets1/2. In sea urchins, Ets1/2 plays a central role in the differentiation of larval skeletogenic mesenchyme cells. In addition, Ets1/2 is suggested to be involved in adult skeletogenesis. Conversely, in starfish, although no skeletogenic cells differentiate during larval development, Ets1/2 is also expressed in the larval mesoderm. Here, we confirmed that the starfish Ets1/2 is indispensable for the differentiation of the larval mesoderm. This result led us to assume that, in the common ancestors of echinoderms, Ets1/2 activates the transcription of distinct gene sets, one for the differentiation of the larval mesoderm and the other for the development of the adult skeleton. Thus, the acquisition of the larval skeleton involved target switching of Ets1/2. Specifically, in the sea urchin lineage, Ets1/2 activated a downstream target gene set for skeletogenesis during larval development in addition to a mesoderm target set. We examined whether this heterochronic activation of the skeletogenic target set was achieved by the molecular evolution of the Ets1/2 transcription factor itself. We tested whether starfish Ets1/2 induced skeletogenesis when injected into sea urchin eggs. We found that, in addition to ectopic induction of mesenchyme cells, starfish Ets1/2 can activate some parts of the skeletogenic pathway in these mesenchyme cells. Thus, we suggest that the nature of the transcription factor Ets1/2 did not change, but rather that some unidentified co-factor(s) for Ets1/2 may distinguish between targets for the larval mesoderm and for skeletogenesis. Identification of the co-factor(s) will be key to understanding the molecular evolution underlying the evolution of the pluteus larvae.


Subject(s)
Echinodermata/genetics , Evolution, Molecular , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-2/genetics , Animals , Cell Differentiation/genetics , Echinodermata/embryology , Echinodermata/growth & development , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Gene Transfer Techniques , In Situ Hybridization , Larva/genetics , Larva/growth & development , Male , Mesoderm/cytology , Mesoderm/metabolism , Phylogeny , Proto-Oncogene Protein c-ets-1/classification , Proto-Oncogene Protein c-ets-1/physiology , Proto-Oncogene Protein c-ets-2/classification , Proto-Oncogene Protein c-ets-2/physiology , Reverse Transcriptase Polymerase Chain Reaction , Sea Urchins/embryology , Sea Urchins/genetics , Sea Urchins/growth & development , Starfish/embryology , Starfish/genetics , Starfish/growth & development
10.
Placenta ; 31(7): 630-40, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20569982

ABSTRACT

Mouse trophoblast stem (TS) cells represent a unique in vitro system that provides an unlimited supply of TS cells for the study of trophoblast differentiation and TS cell self-renewal. Although the mouse transcription factor Ets2 is required for TS cell self-renewal, its role in this and in TS cell differentiation has not been explored fully, partly due to the early lethality of Ets2 null mice. To address this, we developed a novel lentivirus-based system that resulted in efficient Ets2 knockdown in the overwhelming majority of TS cells. This system enables functional studies in TS cells, especially for genes required for TS cell self-renewal because TS cell derivation using gene-knockout embryos for such genes depends on TS cell self-renewal. Using morphological/morphometric criteria and gene expression analysis, we show that the requirement for Ets2 in self-renewal of TS cells cultured in 'stem cell medium' (SCM) involves maintenance of the expression of genes that inhibit TS cell differentiation in SCM, such as Cdx2 and Esrrb, and preservation of the undifferentiated TS cell morphology. During TS cell differentiation caused by Cdx2/Esrrb downregulation, due to either Ets2 knockdown in SCM or culture in differentiation medium (DM), Ets2 is also required for the promotion of trophoblast giant cell (TGC) and junctional zone trophoblast (JZT) differentiation. This TGC differentiation involves Ets2-dependent expression of Hand1, a gene required for the differentiation of all TGC types. This study uncovers new roles for Ets2 in TS cell self-renewal and differentiation and demonstrates the usefulness of this lentivirus system for gene function studies in TS cells.


Subject(s)
Proto-Oncogene Protein c-ets-2/physiology , Stem Cells/physiology , Trophoblasts/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cell Differentiation , Cell Proliferation , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Lentivirus/genetics , Mice , Pregnancy
11.
J Biol Chem ; 285(34): 26417-30, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20554528

ABSTRACT

Different levels of regulation account for the inactivation of MAP kinases by MAPK phosphatases (MKPs), in a cell type- and stimuli-dependent manner. MCF-7 human breast carcinoma cells treated with the phorbol 12-myristate 13-acetate (PMA) suffer growth arrest and show morphological alterations, which depend on the activation of the ERK1/2 MAP kinases. MKP3/DUSP6 and DUSP5 MAP kinase phosphatases, two negative regulators of ERK1/2, were specifically up-regulated in MCF-7 and SKBR3 cells in response to PMA. MKP3 and DUSP5 up-regulation required the prolonged activation of the ERK1/2 pathway, and correlated with the shutdown of this route. MKP3 induction relied on the activation of the Ets2 transcription factor, whereas DUSP5 induction depended on the activation of c-Jun. Diminishing the expression of MKP3 and DUSP5 raised the activation of ERK1/2, and accelerated growth arrest of PMA-treated MCF-7 cells. Conversely, MCF-7 cell lines expressing high levels of MKP3 or DUSP5 did not undergo PMA-triggered growth arrest, displayed a migratory phenotype, and formed colonies in soft agar. We propose that the differential up-regulation of MKP3 by Ets2 and of DUSP5 by c-Jun may converge in similar functional roles for these MAP kinase phosphatases in the growth arrest versus proliferation decisions of breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , Dual Specificity Phosphatase 6/genetics , Dual-Specificity Phosphatases/genetics , Proto-Oncogene Protein c-ets-2/physiology , Proto-Oncogene Proteins c-jun/physiology , Up-Regulation/genetics , Cell Line, Tumor , Cell Proliferation , Female , Humans , Phorbol Esters/pharmacology
12.
Cancer Res ; 70(4): 1323-33, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20145133

ABSTRACT

Tumor-associated macrophages (TAM) are implicated in breast cancer metastasis, but relatively little is known about the underlying genes and pathways that are involved. The transcription factor Ets2 is a direct target of signaling pathways involved in regulating macrophage functions during inflammation. We conditionally deleted Ets in TAMs to determine its function at this level on mouse mammary tumor growth and metastasis. Ets2 deletion in TAMs decreased the frequency and size of lung metastases in three different mouse models of breast cancer metastasis. Expression profiling and chromatin immunoprecipitation assays in isolated TAMs established that Ets2 repressed a gene program that included several well-characterized inhibitors of angiogenesis. Consistent with these results, Ets2 ablation in TAMs led to decreased angiogenesis and decreased growth of tumors. An Ets2-TAM expression signature consisting of 133 genes was identified within human breast cancer expression data which could retrospectively predict overall survival of patients with breast cancer in two independent data sets. In summary, we identified Ets2 as a central driver of a transcriptional program in TAMs that acts to promote lung metastasis of breast tumors.


Subject(s)
Gene Expression Regulation, Neoplastic , Macrophages/metabolism , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Proto-Oncogene Protein c-ets-2/physiology , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/metabolism , Animals , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/mortality , Carcinoma/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Macrophages/pathology , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/mortality , Mice , Mice, Transgenic , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Protein c-ets-2/genetics , Retrospective Studies , Tumor Cells, Cultured
13.
Reprod Biol Endocrinol ; 7: 135, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19939245

ABSTRACT

BACKGROUND: We have previously shown that Matrix metalloproteinase (MMP) -2 is a key-enzyme in early trophoblast invasion and that Protein Kinase A (PKA) increases MMP-2 expression and trophoblast invasion. The aim of this study was to examine MMP -2 regulation by PKA in invasive trophoblasts: JAR choriocarcinoma cell-line and 6-8 w first trimester trophoblasts. METHODS: The effect of Forskolin (PKA) on MMP-2 expression was assessed by Northern Blot and RT-PCR. Possible transcription factors binding to consensus MMP-2 promoter sequences in response to Forskolin, were detected by EMSA binding assay and their expression assessed by western blot analysis. Antisense transfection of relevant transcription factors was performed and the inhibitory effect assessed on MMP-2 expression (RT-PCR), secretion (zymography) and trophoblast invasiveness (transwell migration assay). RESULTS: We found that Forskolin increased MMP-2 mRNA in JAR cells within 24 hours, and induced binding to p53, Ets, C/EBP and AP-2. Transcription factors Ets-2, phospho- p53, C/EBP epsilon, C/EBP lambda and AP-2 alpha bound to their respective binding sequences in response to Forskolin and the expressions of these transcription factors were all elevated in Forskolin- treated cells. Inhibition of Ets-2 and p53 reduced MMP-2 expression, secretion and invasiveness of Forskolin treated cells. CONCLUSION: MMP-2 is regulated by PKA through several binding sites and transcription factors including Ets-2, p53, C/EBP, C/EBP lambda and AP-2 alpha. Ets-2 and p53 mediate cAMP- induced trophoblast invasiveness, through regulation of MMP-2.


Subject(s)
Cyclic AMP/pharmacology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Proto-Oncogene Protein c-ets-2/physiology , Trophoblasts/drug effects , Tumor Suppressor Protein p53/physiology , Binding Sites/drug effects , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cells, Cultured , Colforsin/pharmacology , Gene Expression/drug effects , Humans , Promoter Regions, Genetic/drug effects , Protein Binding/drug effects , Proto-Oncogene Protein c-ets-2/metabolism , Trophoblasts/metabolism , Trophoblasts/physiology , Tumor Suppressor Protein p53/metabolism
14.
Am J Med Genet A ; 149A(10): 2158-65, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19764029

ABSTRACT

Ts65Dn mice have segmental trisomy for orthologs of about half of the genes on human chromosome 21, including Ets2. These mice develop anomalies of the cranial skeleton and thymus that parallel those in Down syndrome. Overexpression of the Ets2 transcription factor gene was posited to be sufficient to produce these craniofacial and thymus deficits in transgenic mice that constitutively overexpress a processed Ets2 transcript under a promiscuous promoter [Sumarsono et al. (1996); Nature 379:534-537; Wolvetang et al. (2003); Hum Mol Genet 12:247-255]. Evaluation of trisomic mice with varying copy numbers of a properly regulated Ets2 gene indicated increased dosage of Ets2 was not sufficient to produce effects on thymus and most of the cranial anomalies seen in Ts65Dn mice. However, mesoderm-derived cranial skeletal elements are significantly more affected in Ts65Dn, Ets2(+/-) mice compared to Ts65Dn littermates suggesting a differential interaction of Ets2-related processes with mesoderm-derived and neural crest-derived formative tissues. Our results support the growing evidence for interactions among multiple genes contributing to developmental perturbations resulting in variation in complex Down syndrome phenotypes.


Subject(s)
Craniofacial Abnormalities/genetics , Down Syndrome/pathology , Mice, Transgenic , Proto-Oncogene Protein c-ets-2/physiology , Thymus Gland/abnormalities , Abnormalities, Multiple/embryology , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Animals , Computer Simulation , Craniofacial Abnormalities/complications , Craniofacial Abnormalities/embryology , Craniofacial Abnormalities/pathology , Disease Models, Animal , Down Syndrome/complications , Down Syndrome/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Models, Anatomic , Phenotype , Proto-Oncogene Protein c-ets-2/genetics , Skull/pathology , Thymus Gland/pathology
15.
Blood ; 114(5): 1123-30, 2009 Jul 30.
Article in English | MEDLINE | ID: mdl-19411629

ABSTRACT

The ras/Raf/Mek/Erk pathway plays a central role in coordinating endothelial cell activities during angiogenesis. Transcription factors Ets1 and Ets2 are targets of ras/Erk signaling pathways that have been implicated in endothelial cell function in vitro, but their precise role in vascular formation and function in vivo remains ill-defined. In this work, mutation of both Ets1 and Ets2 resulted in embryonic lethality at midgestation, with striking defects in vascular branching having been observed. The action of these factors was endothelial cell autonomous as demonstrated using Cre/loxP technology. Analysis of Ets1/Ets2 target genes in isolated embryonic endothelial cells demonstrated down-regulation of Mmp9, Bcl-X(L), and cIAP2 in double mutants versus controls, and chromatin immunoprecipitation revealed that both Ets1 and Ets2 were loaded at target promoters. Consistent with these observations, endothelial cell apoptosis was significantly increased both in vivo and in vitro when both Ets1 and Ets2 were mutated. These results establish essential and overlapping functions for Ets1 and Ets2 in coordinating endothelial cell functions with survival during embryonic angiogenesis.


Subject(s)
Apoptosis/genetics , Embryonic Development/genetics , Endothelial Cells/cytology , Gene Expression Regulation, Developmental/physiology , Neovascularization, Physiologic/genetics , Proto-Oncogene Protein c-ets-1/physiology , Proto-Oncogene Protein c-ets-2/physiology , Animals , Blood Vessels/embryology , Blood Vessels/ultrastructure , Cell Survival/genetics , Chimera , Edema/embryology , Edema/genetics , Embryo Transfer , Fetal Death/genetics , Fetal Death/pathology , Fetal Diseases/genetics , Fetal Diseases/pathology , Gene Expression Regulation, Developmental/genetics , Genetic Vectors/genetics , Genetic Vectors/pharmacology , Hemorrhage/embryology , Hemorrhage/genetics , Homozygote , Mice , Mice, Knockout , Phenotype , Proto-Oncogene Protein c-ets-1/deficiency , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-2/deficiency , Proto-Oncogene Protein c-ets-2/genetics
16.
Plast Reconstr Surg ; 123(2 Suppl): 83S-93S, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19182667

ABSTRACT

BACKGROUND: Mechanotransduction underpins the homeostasis of musculoskeletal tissues, including cranial sutures. Intracellular calcium, [Ca 2+]ic, and protein phosphorylation are two intermediate variables in signal relay during mechanotransduction. This project establishes a chain of cause and effect, linking cellular strain to substrate phosphorylation, and identifies the agent and target sites of phosphorylation. METHODS: Cyclic tensile force (0.5 N at 1 Hz) was applied to 1-day-old rat sagittal sutures. [Ca 2+]ic was measured by FURA-2. Ets-2 phosphorylation by CaMKII was tested using Western blot autoradiography. Peptide array was constructed to determine the precise sites of phosphorylation. The results were confirmed with mass spectroscopy and Western blots using phospho-specific antibodies. RESULTS: [Ca 2+]ic increased rapidly in response to tensile stress. In the presence of Ca2+, CaMKII caused Ets-2 phosphorylation. Of the three possible sites for phosphorylation of Ets-2 by CaMKII, RVPS, FESF, RLSS, Serine 246, 310, and 313 were the targets. Furthermore, the contiguous sequence modified this effect. Mass spectroscopy showed 80 Da (molecular weight of phosphate group, -PO3) right shifts consistent with phosphorylation. There was cytosolic translocation of Ets-2 on tensile deformation of suture cells. CaMKII binding of Ets-2 occurred within 30 minutes after the onset of tensile strain. CONCLUSIONS: Cranial suture cells can respond to tensile forces by increasing [Ca 2+]ic, which causes CaMKII to phosphorylate Ets-2, thus altering Ets-2 binding to its downstream promoters. Of note, Ets-2 is at the intersection of three key pathways important in craniosynostosis: fibroblast growth factor-2, transforming growth factor-beta, and mechanotransduction.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cranial Sutures/metabolism , Musculoskeletal System/metabolism , Proto-Oncogene Protein c-ets-2/physiology , Tensile Strength , Animals , Animals, Newborn , Homeostasis , Models, Animal , Models, Biological , Phosphorylation , Proto-Oncogene Protein c-ets-2/metabolism , Rats
17.
Blood ; 113(14): 3337-47, 2009 Apr 02.
Article in English | MEDLINE | ID: mdl-19168790

ABSTRACT

ETS2 and ERG are transcription factors, encoded on human chromosome 21 (Hsa21), that have been implicated in human cancer. People with Down syndrome (DS), who are trisomic for Hsa21, are predisposed to acute megakaryoblastic leukemia (AMKL). DS-AMKL blasts harbor a mutation in GATA1, which leads to loss of full-length protein but expression of the GATA-1s isoform. To assess the consequences of ETS protein misexpression on megakaryopoiesis, we expressed ETS2, ERG, and the related protein FLI-1 in wild-type and Gata1 mutant murine fetal liver progenitors. These studies revealed that ETS2, ERG, and FLI-1 facilitated the expansion of megakaryocytes from wild-type, Gata1-knockdown, and Gata1s knockin progenitors, but none of the genes could overcome the differentiation block characteristic of the Gata1-knockdown megakaryocytes. Although overexpression of ETS proteins increased the proportion of CD41(+) cells generated from Gata1s-knockin progenitors, their expression led to a significant reduction in the more mature CD42 fraction. Serial replating assays revealed that overexpression of ERG or FLI-1 immortalized Gata1-knockdown and Gata1s knockin, but not wild-type, fetal liver progenitors. Immortalization was accompanied by activation of the JAK/STAT pathway, commonly seen in megakaryocytic malignancies. These findings provide evidence for synergy between alterations in GATA-1 and overexpression of ETS proteins in aberrant megakaryopoiesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , GATA1 Transcription Factor/physiology , Hematopoietic Stem Cells/pathology , Oncogene Proteins/physiology , Proto-Oncogene Protein c-ets-2/physiology , Thrombopoiesis/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Embryo, Mammalian , Fetus/metabolism , Fetus/physiology , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Liver/embryology , Liver/metabolism , Liver/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Proto-Oncogene Protein c-ets-2/genetics , Proto-Oncogene Protein c-ets-2/metabolism , Thrombopoiesis/physiology , Transcription Factors , Transcriptional Regulator ERG
18.
Biol Reprod ; 79(1): 115-24, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18322277

ABSTRACT

Distal-less 3 (DLX3), a homeodomain transcription factor required for placental development in the mouse, modestly transactivates hCG-alpha subunit gene (hCGA) expression in human choriocarcinoma cells. Because hCG and interferon-tau (IFNT) are expressed in trophectoderm of primates and ruminants, respectively, we have tested the hypothesis that DLX3 regulates the genes for IFNT (IFNT). A bovine IFNT1 promoter (-457 to +66), linked to a luciferase (luc) reporter, was transactivated approximately 20-fold by overexpressing DLX3 in human JAr cells. Elimination of a potential DLX3-binding site (-54 GATAATGAG -46) by either truncation or mutagenesis abolished this effect. A sequence (-59 to -44) encompassing this site bound DLX3 specifically. Coexpression of DLX3 and ETS2, which is known to be a key regulator of IFNT expression, increased reporter activity by more than 250-fold, whereas deletion of the established ETS2 site (-79 to -70) eliminated the ability of DLX3 to transactivate the gene. Conversely, mutation of the DLX3 site significantly reduced the transactivational effects of ETS2. Both DLX3 and ETS2 are coexpressed in JAr cells and in an IFNT-producing, bovine trophoblast cell line, CT-1. The two can be immunoprecipitated together as a complex from CT-1 cells, and RNAi-mediated, partial knockdown of DLX3 expression reduced the production of IFNT by approximately 50+. Together, these results suggest that DLX3 has a central role in controlling IFNT gene expression by associating with ETS2 on the IFNT promoter.


Subject(s)
Cattle/genetics , Homeodomain Proteins/metabolism , Interferon Type I/genetics , Pregnancy Proteins/genetics , Proto-Oncogene Protein c-ets-2/metabolism , Transcription Factors/metabolism , Transcription Factors/physiology , Animals , Base Sequence , Binding Sites/genetics , Cells, Cultured , Genes, Reporter , Homeodomain Proteins/physiology , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Protein c-ets-2/physiology , Sheep , Transcriptional Activation , Transfection
19.
Leukemia ; 22(3): 521-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18094719

ABSTRACT

Acute myeloid leukemia (AML) in Down syndrome (DS) children has several unique features including a predominance of the acute megakaryocytic leukemia (AMkL) phenotype, higher event-free survivals compared to non-DS children using cytosine arabinoside (ara-C)/anthracycline-based protocols and a uniform presence of somatic mutations in the X-linked transcription factor gene, GATA1. Several chromosome 21-localized transcription factor oncogenes including ETS2 may contribute to the unique features of DS AMkL. ETS2 transcripts measured by real-time RT-PCR were 1.8- and 4.1-fold, respectively, higher in DS and non-DS megakaryoblasts than those in non-DS myeloblasts. In a doxycycline-inducible erythroleukemia cell line, K562pTet-on/ETS2, induction of ETS2 resulted in an erythroid to megakaryocytic phenotypic switch independent of GATA1 levels. Microarray analysis of doxycycline-induced and doxycycline-uninduced cells revealed an upregulation by ETS2 of cytokines (for example, interleukin 1 and CSF2) and transcription factors (for example, TAL1), which are key regulators of megakaryocytic differentiation. In the K562pTet-on/ETS2 cells, ETS2 induction conferred differences in sensitivities to ara-C and daunorubicin, depending on GATA1 levels. These results suggest that ETS2 expression is linked to the biology of AMkL in both DS and non-DS children, and that ETS2 acts by regulating expression of hematopoietic lineage and transcription factor genes involved in erythropoiesis and megakaryopoiesis, and in chemotherapy sensitivities.


Subject(s)
Gene Expression Regulation, Leukemic/physiology , Leukemia, Myeloid/etiology , Neoplasm Proteins/physiology , Proto-Oncogene Protein c-ets-2/physiology , Acute Disease , Cell Differentiation/genetics , Child , Chromosomes, Human, Pair 21/genetics , Cytarabine/pharmacology , Daunorubicin/pharmacology , Down Syndrome/complications , Drug Resistance, Neoplasm/genetics , Erythroid Precursor Cells/metabolism , Erythropoiesis/genetics , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/physiology , Gene Dosage , Gene Expression Regulation, Leukemic/genetics , Genetic Predisposition to Disease , Humans , K562 Cells/drug effects , K562 Cells/metabolism , Leukemia, Megakaryoblastic, Acute/etiology , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/genetics , Megakaryocytes/drug effects , Megakaryocytes/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Proto-Oncogene Mas , Thrombopoiesis/genetics
20.
Biochem Biophys Res Commun ; 348(2): 768-74, 2006 Sep 22.
Article in English | MEDLINE | ID: mdl-16890195

ABSTRACT

Autoimmune regulator (AIRE) directs the expression of self-antigens in thymus. Defects in AIRE gene cause an organ-specific autoimmune disease called autoimmune polyendocrinopathy candidiasis ectodermal dystrophy (APECED). AIRE protein is mainly expressed in thymic medullary epithelial cells, thus implying a strict control over its expression pattern. To date, only limited information is available on mechanisms responsible for the regulation of AIRE gene. Here, we show that Ets transcription factor family members Ets-1, Ets-2, and ESE-1 have positive effect on AIRE transcription. Site-directed mutagenesis and transfection studies revealed that two of the three Ets binding sites in AIRE promoter are functional and this finding has been confirmed by the electrophoretic mobility shift assay. The AIRE promoter activity could be stimulated by phorbol myristate acetate (PMA) and this activation was further enhanced by Ets transcription factors. Our results demonstrate for the first time that AIRE gene is a downstream target for the Ets family of transcription factors.


Subject(s)
Promoter Regions, Genetic , Proto-Oncogene Proteins c-ets/physiology , Transcription Factors/genetics , Base Sequence , Binding Sites , DNA-Binding Proteins/physiology , Electrophoretic Mobility Shift Assay , HeLa Cells , Humans , Molecular Sequence Data , Promoter Regions, Genetic/drug effects , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Protein c-ets-2/physiology , Proto-Oncogene Proteins/physiology , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors/physiology , AIRE Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...