Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nucleic Acids Res ; 40(18): 8993-9007, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22844087

ABSTRACT

Rex1/Zfp42 is a Yy1-related zinc-finger protein whose expression is frequently used to identify pluripotent stem cells. We show that depletion of Rex1 levels notably affected self-renewal of mouse embryonic stem (ES) cells in clonal assays, in the absence of evident differences in expression of marker genes for pluripotency or differentiation. By contrast, marked differences in expression of several endogenous retroviral elements (ERVs) were evident upon Rex1 depletion. We demonstrate association of REX1 to specific elements in chromatin-immunoprecipitation assays, most strongly to muERV-L and to a lower extent to IAP and musD elements. Rex1 regulates muERV-L expression in vivo, as we show altered levels upon transient gain-and-loss of Rex1 function in pre-implantation embryos. We also find REX1 can associate with the lysine-demethylase LSD1/KDM1A, suggesting they act in concert. Similar to REX1 binding to retrotransposable elements (REs) in ES cells, we also detected binding of the REX1 related proteins YY1 and YY2 to REs, although the binding preferences of the two proteins were slightly different. Altogether, we show that Rex1 regulates ERV expression in mouse ES cells and during pre-implantation development and suggest that Rex1 and its relatives have evolved as regulators of endogenous retroviral transcription.


Subject(s)
Embryonic Stem Cells/metabolism , Endogenous Retroviruses/genetics , Transcription Factors/metabolism , Animals , Biomarkers/metabolism , Cell Line , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Endogenous Retroviruses/metabolism , Gene Expression Regulation , Histone Demethylases , Mice , Oxidoreductases, N-Demethylating/metabolism , RNA, Messenger/metabolism , Retroelements , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , YY1 Transcription Factor/metabolism
2.
Curr Biol ; 11(10): 793-7, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11378392

ABSTRACT

Fibroblast growth factors (FGFs) mediate cell growth, differentiation, migration, and morphogenesis by binding to the extracellular domain of cell surface receptors, triggering receptor tyrosine phosphorylation and signal transduction [1-5]. FGF homologous factors (FHFs) were discovered within vertebrate DNA sequence databases by virtue of their sequence similarity to FGFs [3, 6, 7], but the mechanism of FHF action has not been reported. We show here that FHF-1 is associated with the MAP kinase (MAPK) scaffold protein Islet-Brain-2 (IB2) [8] in the brain and in specific cell lines. FHF/IB2 interaction is highly specific, as FHFs do not bind to the related scaffold protein IB1(JIP-1b) [9, 10], nor can FGF-1 bind to IB2. We further show that FHFs enable IB2 to recruit a specific MAPK in transfected cells, and our data suggest that the scaffolds IB1 and IB2 have different MAPK specificities. Hence, FHFs are intracellular components of a tissue-specific protein kinase signaling module.


Subject(s)
Fibroblast Growth Factors/physiology , Signal Transduction/physiology , Animals , Fibroblast Growth Factors/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , Protein Binding , Two-Hybrid System Techniques
3.
Mol Cell Biol ; 14(2): 1122-36, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8289793

ABSTRACT

Expression of Oct4 in embryonic stem cells is controlled by a distal upstream stem cell-specific enhancer that is deactivated during retinoic acid (RA)-induced differentiation by an indirect mechanism not involving binding of RA receptors (H. Okazawa, K. Okamoto, F. Ishino, T. Ishino-Kaneko, S. Takeda, Y. Toyoda, M. Muramatsu, and H. Hamada, EMBO J. 10:2997-3005, 1991). Here we report that in RA-treated P19 embryonal carcinoma cells the Oct4 promoter is also subject to negative regulation by RA. The minimal Oct4 promoter sequence mediating repression consists of a promoter-proximal sequence containing a GC-rich SP1 consensus-like sequence and several hormone response element half-sites that can be arranged into direct repeats with different spacing. The GC box binds a nuclear factor that is invariably present in undifferentiated and RA-treated differentiated P19 cells. By contrast, the hormone response element-containing sequence binds factors that are induced following RA treatment. Mutational analysis and competition experiments show that the functional entity binding the RA-induced factor is a direct repeat sequence with a spacing of one nucleotide, previously shown to be a binding site for COUP transcription factors (COUP-TFs). Cotransfected orphan receptors COUP-TF1, ARP-1, and EAR-2 were able to repress the activity of Oct4 promoter-driven reporters in P19 EC cells, albeit with different efficiencies. Furthermore, the negative transcriptional effect of COUP-TFs is dominant over the activating effect of the Oct4 embryonic stem cell-specific enhancer. These results show that negative regulation of Oct4 expression during RA-induced differentiation of embryonic stem cells is controlled by two different mechanisms, including deactivation of the embryonic stem cell-specific enhancer and promoter silencing by orphan nuclear hormone receptors.


Subject(s)
DNA-Binding Proteins/genetics , Promoter Regions, Genetic/drug effects , Transcription Factors/genetics , Tretinoin/pharmacology , Animals , Base Sequence , Binding Sites , Cell Differentiation , Cell Line , Consensus Sequence , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Genes, Regulator/drug effects , Genomic Library , Mice , Mice, Inbred CBA , Mice, Inbred Strains , Molecular Sequence Data , Octamer Transcription Factor-3 , Oligodeoxyribonucleotides , Repetitive Sequences, Nucleic Acid , Restriction Mapping , Sequence Homology, Nucleic Acid , Spleen/metabolism , Stem Cells/metabolism , Transcription, Genetic , Transfection
4.
Placenta ; 55: 21-28, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28623969

ABSTRACT

INTRODUCTION: Suspected preterm labour occurs in around 9% of pregnancies. However, almost two-thirds of women admitted for threatened preterm labour ultimately deliver at term and are considered risk-free for fetal development. METHODS: We examined placental and umbilical cord blood samples from preterm or term deliveries after threatened preterm labour as well as term deliveries without threatened preterm labour. We quantitatively analysed the mRNA expression of inflammatory markers (IL6, IFNγ, and TNFα) and modulators of angiogenesis (FGF2, PGF, VEGFA, VEGFB, and VEGFR1). RESULTS: A total of 132 deliveries were analysed. Preterm delivery and term delivery after suspected preterm labour groups showed similar increases in TNFα expression compared with the term delivery control group in umbilical cord blood samples. Placental samples from preterm and term deliveries after suspected preterm labour exhibited significantly increased expression of TNFα and IL6 and decreased expression of IFNγ. Suspected preterm labour was also associated with altered expression of angiogenic factors, although not all differences reached statistical significance. DISCUSSION: We found gene expression patterns indicative of inflammation in human placentas after suspected preterm labour regardless of whether the deliveries occurred preterm or at term. Similarly, a trend towards altered expression of angiogeneic factors was not limited to preterm birth. These findings suggest that the biological mechanisms underlying threatened preterm labour affect pregnancies independently of gestational age at birth.


Subject(s)
Inflammation/metabolism , Neovascularization, Physiologic , Obstetric Labor, Premature/metabolism , Placenta/metabolism , Adult , Biomarkers/blood , Female , Fetal Blood/metabolism , Gene Expression , Humans , Pregnancy , Prospective Studies , Young Adult
5.
Mech Dev ; 36(1-2): 75-86, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1723621

ABSTRACT

Expression of kFGF, which belongs to the family of fibroblast growth factor genes, is restricted to undifferentiated embryonal carcinoma and embryonic stem cells. Stem cell specific expression of kFGF is controlled by a distally localized enhancer, conferring both positive and negative regulation to the kFGF and tk promoters. This enhancer contains a consensus octamer binding sequence that controls positive regulation in EC and ES cells. The octamer sequence binds Oct1 and Oct4 in nuclear extracts from undifferentiated EC cells, while only Oct1 is bound in nuclear extracts from RA differentiated cells. These results suggest that the kFGF gene is a target for positive regulation by Oct4 and implicate Oct4 as target for regulation by the retinoic acid receptors.


Subject(s)
Fibroblast Growth Factors , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins/genetics , Regulatory Sequences, Nucleic Acid , Transcription Factors , Animals , Binding Sites , Blotting, Northern , Cell Differentiation , Cell Line , DNA/metabolism , DNA, Recombinant , DNA-Binding Proteins/metabolism , Dimethyl Sulfoxide/metabolism , Down-Regulation , Embryonal Carcinoma Stem Cells , Enhancer Elements, Genetic , Fibroblast Growth Factor 4 , Gene Expression Regulation , Humans , Mice , Neoplastic Stem Cells/cytology , Octamer Transcription Factor-3 , Promoter Regions, Genetic , RNA , RNA, Messenger , Transfection , Tretinoin
6.
Mech Dev ; 47(1): 81-97, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7947324

ABSTRACT

We have isolated the murine homologs of the members of the COUP-family of steroid hormone receptors, COUP-TF1, ARP-1 and EAR2. The proteins encoded by the murine genes appeared to be highly conserved when compared to their human counterparts. The expression of COUP-TF1 and ARP-1 was induced during differentiation of P19 embryonal carcinoma (EC) cells into derivatives of all three germ layers. Retinoic acid (RA) treatment rapidly induced expression of both genes, while other methods of differentiation were less effective. Undifferentiated P19 cells were found to express EAR2 mRNA and the expression level was only slightly elevated by RA-treatment. In addition, we analyzed the expression in P19 cells of three members of the retinoid X receptor (RXR) family, which have been shown to heterodimerize with members of the COUP-family. During RA mediated differentiation of P19 cells, RXR alpha expression was induced while RXR beta expression was not modulated and RXR gamma expression was down regulated. Gel shift analysis revealed that in P19 cells the members of the COUP-family comprise the major portion of proteins binding to a RA-responsive direct repeat of the consensus steroid hormone receptor binding half site (AGGTCA) spaced by one nucleotide (DR + 1). The members of the COUP-family appeared to down regulate RA-induced activation of RA-response element-containing reporter constructs in a promoter context-dependent manner. The expression patterns of COUP-TF1, ARP-1 and EAR2 during development were investigated by in situ hybridization. In agreement with the results obtained in vitro, the three genes appeared to be expressed in tissues derived from all three germ layers. However, COUP-TF1 and ARP-1 were found to be expressed predominantly in the developing central nervous system in mutually exclusive domains. Furthermore, strong ARP-1 expression was detected in lung and kidney. Our data strongly suggest an important role for the members of the COUP-family in the hormonal control of gene expression regulating embryogenesis.


Subject(s)
DNA-Binding Proteins/genetics , Receptors, Steroid/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , COUP Transcription Factor I , COUP Transcription Factor II , COUP Transcription Factors , Carcinoma, Embryonal/chemistry , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/pathology , Cloning, Molecular , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , DNA-Binding Proteins/analysis , Gene Expression Regulation, Neoplastic , In Situ Hybridization , Mice , Molecular Sequence Data , RNA, Messenger/analysis , RNA, Messenger/genetics , Receptors, Retinoic Acid/analysis , Receptors, Retinoic Acid/genetics , Receptors, Steroid/analysis , Retinoid X Receptors , Transcription Factors/analysis , Tretinoin/pharmacology , Tumor Cells, Cultured
7.
Stem Cell Res ; 7(1): 1-16, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21530438

ABSTRACT

Rex-1/Zfp42 displays a remarkably restricted pattern of expression in preimplantation embryos, primary spermatocytes, and undifferentiated mouse embryonic stem (ES) cells and is frequently used as a marker gene for pluripotent stem cells. To understand the role of Rex-1 in selfrenewal and pluripotency, we used Rex-1 association as a measure to identify potential target genes, and carried out chromatin-immunoprecipitation assays in combination with gene specific primers to identify genomic targets Rex-1 associates with. We find association of Rex-1 to several genes described previously as bivalently marked regulators of differentiation and development, whose repression in mouse embryonic stem (ES) cells is Polycomb Group-mediated, and controlled directly by Ring1A/B. To substantiate the hypothesis that Rex-1 contributes to gene regulation by PcG, we demonstrate interactions of Rex-1 and YY2 (a close relative of YY1) with Ring1 proteins and the PcG-associated proteins RYBP and YAF2, in line with interactions reported previously for YY1. We also demonstrate the presence of Rex-1 protein in both trophectoderm and Inner Cell Mass of the mouse blastocyst and in both ES and in trophectoderm stem (TS) cells. In TS cells, we were unable to demonstrate association of Rex-1 to the genes it associates with in ES cells, suggesting that association may be cell-type specific. Rex-1 might fine-tune pluripotency in ES cells by modulating Polycomb-mediated gene regulation.


Subject(s)
Embryonic Stem Cells/physiology , Gene Expression Regulation , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Repressor Proteins/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Embryonic Stem Cells/metabolism , Female , Genes, Homeobox , Humans , Mice , Polycomb-Group Proteins , Pregnancy , Rabbits , Repressor Proteins/genetics , Transfection
8.
Mol Biol Rep ; 21(3): 129-40, 1995.
Article in English | MEDLINE | ID: mdl-8832901

ABSTRACT

The stem cell-specific factor Oct-4 is expressed in undifferentiated embryonal carcinoma and embryonic stem cells and is quickly down regulated upon RA-induced differentiation. Irrespective of the direction of differentiation, Oct-4 repression in P19 EC cells requires treatment with high doses of either all-trans or 9-cis RA. Unlike in P19 cells, no RA-induced down regulation of Oct-4 expression is observed in the P19-derived RA-resistant RAC65 cells. However, in these cells Oct-4 promoter repression can be rescued in a RA-dependent manner by cotransfection of RAR alpha 2 or RAR beta 2 but not RARr gamma 1, matching previously reported transactivation properties of these receptor types. In the vicinity of the transcription initiation site of the Oct-4 gene, three Hormone Response Element HRE half sites are present which are arranged as direct repeats with different spacing. In vitro translated RAR and RXR proteins bind to this HRE as heterodimers with low affinity, in such a way that all three HRE half sites contribute to complex formation. Although P19 EC cells contain weak binding activity interacting with the Oct-4 promoter HRE, strong binding activity is observed in nuclear extracts from RA-treated P19 cells. This binding activity was shown to correspond to COUP-TFs but not nuclear RA receptors. Moreover, the presence of these binding factors in nuclear extracts corresponds to silencing of Oct-4 expression. These results implicate RA and the action of its nuclear receptors in silencing Oct-4 expression upon differentiation of EC cells. The observed silencing is most likely not exerted by direct binding of RARs to the Oct-4 proximal promoter HRE. Our results support models in which different nuclear receptor complexes sequentially occupy different sites in the Oct-4 promoter HRE to silence Oct-4 expression during RA-induced differentiation.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation/physiology , Neoplastic Stem Cells/cytology , Transcription Factors , Cell Differentiation , Cell Extracts , Cell Nucleus/metabolism , DNA/metabolism , Embryonal Carcinoma Stem Cells , Gene Expression Regulation/drug effects , Humans , Neoplastic Stem Cells/physiology , Octamer Transcription Factor-3 , Promoter Regions, Genetic/genetics , RNA, Messenger/analysis , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Repetitive Sequences, Nucleic Acid/genetics , Stem Cells/cytology , Stem Cells/metabolism , Transfection , Tretinoin/pharmacology
9.
Nucleic Acids Res ; 18(11): 3195-202, 1990 Jun 11.
Article in English | MEDLINE | ID: mdl-2113273

ABSTRACT

The jun and fos gene families encode DNA binding proteins involved in transcriptional regulation of genes containing a TPA responsive element (TRE). To study their role in gene regulation during early mammalian development, expression and transcription regulatory properties of their gene products were investigated during retinoic acid (RA) induced differentiation of P19 embryonal carcinoma (EC) cells. Our results show, that c-jun is expressed at low but detectable levels in undifferentiated P19 EC cells and at elevated levels in its RA differentiated derivatives, corresponding with increased expression of Jun and TRE binding activity. Jun D is constitutively expressed at constant levels in both undifferentiated and differentiated P19 cells, while jun B and c-fos are not expressed. Addition of TPA to undifferentiated P19 cells does not result in induction of c-jun, jun B and c-fos, while these genes are transiently induced in RA-differentiated P19 cells. In addition, TPA treatment resulted in expression of Fos and Jun protein in RA-differentiated, but not in undifferentiated P19 cells. Addition of TPA to P19 EC cells expressing low levels of TRE binding proteins is neither followed by transcriptional activation of the TRE reporter gene nor by induction of c-jun, previously shown to be autoregulated by its own gene product. By contrast, in P19 cells differentiated by RA that contain elevated levels of TRE binding proteins, TRE dependent transcription is enhanced upon TPA treatment.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/cytology , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Transcription Factors/genetics , Animals , Blotting, Northern , Cell Differentiation , Embryonal Carcinoma Stem Cells , Mice , Neoplastic Stem Cells/metabolism , Plasmids , Proto-Oncogene Proteins c-fos , Proto-Oncogene Proteins c-jun , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic , Tretinoin/pharmacology
10.
Development ; 127(23): 5093-100, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11060235

ABSTRACT

The products of the Polycomb group (PcG) of genes act as transcriptional repressors involved in the maintenance of homeotic gene expression patterns throughout development, from flies to mice. Biochemical and molecular evidence suggests that the mouse Ring1A gene is a member of the PcG of genes. However, genetic evidence is needed to establish PcG function for Ring1A, since contrary to all other murine PcG genes, there is no known Drosophila PcG gene encoding a homolog of the Ring1A protein. To study Ring1A function we have generated a mouse line lacking Ring1A and mouse lines overexpressing Ring1A. Both Ring1A(-/-)and Ring1A(+/-) mice show anterior transformations and other abnormalities of the axial skeleton, which indicates an unusual sensitivity of axial skeleton patterning to Ring1A gene dosage. Ectopic expression of Ring1A also results in dose-dependent anterior transformations of vertebral identity, many of which, interestingly, are shared by Ring1A(-/-) mice. In contrast, the alterations of Hox gene expression observed in both type of mutant mice are subtle and involve a reduced number of Hox genes. Taken together, these results provide genetic evidence for a PcG function of the mouse Ring1A gene.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Repressor Proteins/physiology , Ribs/abnormalities , Spine/abnormalities , Transcription Factors/physiology , Zebrafish Proteins , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Homeodomain Proteins/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutagenesis , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Transcription Factors/genetics , Transcription Factors/metabolism
11.
EMBO J ; 16(19): 5930-42, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9312051

ABSTRACT

In Drosophila, the products of the Polycomb group (Pc-G) of genes act as chromatin-associated multimeric protein complexes that repress expression of homeotic genes. Vertebrate Pc-G homologues have been identified, but the nature of the complexes they form and the mechanisms of their action are largely unknown. The Polycomb homologue M33 is implicated in mesoderm patterning in the mouse and here we show that it acts as a transcriptional repressor in transiently transfected cells. Furthermore, we have identified two murine proteins, Ring1A and Ring1B, that interact directly with the repressor domain of M33. Ring1A and Ring1B display blocks of similarity throughout their sequences, including an N-terminal RING finger domain. However, the interaction with M33 occurs through a region at the C-terminus. Ring1A represses transcription through sequences not involved in M33 binding. Ring1A protein co-localizes in nuclear domains with M33 and other Pc-G homologues, such as Bmi1. The expression of Ring1A at early stages of development is restricted to the neural tube, whereas M33 is expressed ubiquitously. Within the neural tube, Ring1A RNA is located at the rhombomere boundaries of the hindbrain. Taken together, these data suggest that Ring1A may contribute to a tissue-specific function of Pc-G-protein complexes during mammalian development.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins , Insect Proteins/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Zinc Fingers , 3T3 Cells , Amino Acid Sequence , Animals , COS Cells , DNA, Complementary/chemistry , DNA, Complementary/isolation & purification , Drosophila , Gene Expression Regulation, Developmental , In Situ Hybridization , Insect Proteins/chemistry , Mice , Molecular Sequence Data , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Repressor Proteins/chemistry
12.
Dev Biol ; 142(2): 406-13, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2257974

ABSTRACT

Embryonic stem (ES) cells, derived in culture directly from the inner cells mass (ICM) of blastocysts, more closely resemble their embryonic counterparts than the more commonly used embryonal carcinoma (EC) cells derived from teratocarcinomas. In view of the potential role of growth factors in early development, we have now followed changes in the expression of transforming growth factor beta (in particular TGF beta 1, beta 3, beta 4), platelet-derived growth factor (PDGF-A, PDGF-B) and insulin-like growth factor (IGF II) during the differentiation of ES cells in monolayer. When maintained in medium conditioned by Buffalo rat liver cells (BRL-CM) to inhibit differentiation, ES cells expressed 2.5 and 1.8 kb transcripts for TGF beta 1, as well as transcripts for TGF beta 4, PDGF-A, and low levels of PDGF-B, but not TGF beta 3 or IGF II. After formation of parietal endoderm-like cells by addition of retinoic acid (RA) to BRL-CM, the 1.8-kb transcript of TGF beta 1 and PDGF-A expression were reduced, IGF II mRNA and a single TGF beta 3 transcript of 3.8 kb were induced while PDGF-B and TGF beta 4 remained virtually unchanged. By contrast, in ES cells induced to differentiate by the absence of BRL-CM, unusual transcripts for TGF beta 3 of 3.0 and 6.0 kb became detectable and PDGF-B expression increased. The changes in growth factor expression in ES cells are compared with those in F9 and P19 EC cells induced to differentiate in monolayer by RA.


Subject(s)
Blastocyst/cytology , Growth Substances/biosynthesis , Stem Cells/physiology , Animals , Cell Differentiation , Cells, Cultured , Gene Expression , Growth Substances/genetics , Mice , Platelet-Derived Growth Factor/biosynthesis , Platelet-Derived Growth Factor/genetics , RNA, Messenger/biosynthesis , Somatomedins/biosynthesis , Somatomedins/genetics , Stem Cells/metabolism , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics
SELECTION OF CITATIONS
SEARCH DETAIL