Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
2.
BMC Res Notes ; 8: 676, 2015 Nov 14.
Article in English | MEDLINE | ID: mdl-26576650

ABSTRACT

BACKGROUND: Gastric cancers present late in life with advanced disease and carry a poor prognosis. Polo-like Kinase 1 (PLK1) is a mitotic kinase with regulatory functions during G2/M and mitosis in the cell cycle. In mammalian cells, there is an intricate co-regulatory relationship between PLK1 and the forkhead transcription factor FOXM1. It has been demonstrated that individually either PLK1 or FOXM1 expression predicts poorer survival. However, the co-expression of both of these markers in gastric adenocarcinomas has not been reported previously. METHODS: We aimed to assess the expression of PLK1 and FOXM1 in Gastric adenocarcinomas in a Western Population, to examine whether there is a relationship of PLK1 to FOXM1 in cancer samples. We assess both the protein and mRNA expression in this patient population by Tissue Microarray immunohistochemistry and RT-PCR. RESULTS: Immunohistochemistry was performed on biopsy samples from 79 patients with gastric cancer. Paired normal controls were available in 47 patients. FOXM1 expression was significantly associated with gastric adenocarcinoma (p = 0.001). PLK1 and FOXM1 co-expression was demonstrated in 6/8 (75 %) tumours when analysed by RT-PCR. FOXM1 is overexpressed in a large proportion of gastric carcinomas at the protein level and FOXM1 and PLK1 are concomitantly overexpressed at the mRNA level in this cancer type. CONCLUSIONS: This study has demonstrated that FOXM1 and its target gene PLK1 are coordinately overexpressed in a proportion of gastric adenocarcinomas. This suggests that chemotherapeutic treatments that target this pathway may be of clinical utility.


Subject(s)
Adenocarcinoma/metabolism , Cell Cycle Proteins/metabolism , Forkhead Transcription Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Stomach Neoplasms/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Aged , Cell Cycle Proteins/genetics , Female , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Humans , Male , Middle Aged , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Polo-Like Kinase 1
3.
Oncogenesis ; 4: e167, 2015 Sep 07.
Article in English | MEDLINE | ID: mdl-26344694

ABSTRACT

The forkhead transcription factor FOXK2 has recently been implicated in cancer cell proliferation and survival, but a role in cancer chemotherapeutic drug resistance has hitherto not been explored. Here we demonstrate that FOXK2 has a central role in mediating the cytotoxic drug response in breast cancer. Clonogenic and cell viability assays showed that enhanced FOXK2 expression sensitizes MCF-7 breast cancer cells to paclitaxel or epirubicin treatment, whereas FOXK2 depletion by small interfering RNAs (siRNAs) confers drug resistance. Our data also showed that the activation of the tumour suppressor FOXO3a by paclitaxel and epirubicin is mediated through the induction of FOXK2, as depletion of FOXK2 by siRNA limits the induction of FOXO3a by these drugs in MCF-7 cells. Chromatin immunoprecipitation (ChIP) analysis showed that in response to drug treatment, FOXK2 accumulates and binds to the proximal FOXO3a promoter region in MCF-7 cells. Furthermore, we also uncovered that FOXK2 is deregulated and, therefore, can express at high levels in the nucleus of both the paclitaxel and epirubicin drug-resistant MCF-7 cells. Our results showed that ectopically overexpressed FOXK2 accumulates in the nuclei of drug-resistant MCF-7 cells but failed to be recruited to target genes, including FOXO3a. Crucially, we found that FOXO3a is required for the anti-proliferative and epirubicin-induced cytotoxic function of FOXK2 in MCF-7 cells by sulphorhodamine and clonogenic assays. The physiological importance of the regulation of FOXO3a by FOXK2 is further confirmed by the significant correlations between FOXO3a and FOXK2 expression in breast carcinoma patient samples. Further survival analysis also reveals that high nuclear FOXK2 expression significantly associates with poorer clinical outcome, particularly in patients who have received conventional chemotherapy, consistent with our finding that FOXK2 is deregulated in drug-resistant cells. In summary, our results suggest that paclitaxel and epirubicin target the FOXK2 to modulate their cytotoxicity and deregulated FOXK2 confers drug resistance.

4.
Br J Cancer ; 107(10): 1766-75, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23037713

ABSTRACT

BACKGROUND: The transcription factor FOXM1 is an important regulator of the cell cycle through controlling periodic gene expression during the G2 and M phases. One key target for FOXM1 is the gene encoding the protein kinase PLK1 and PLK1 itself acts in a positive feedback loop to phosphorylate and activate FOXM1. Both FOXM1 and PLK1 have been shown to be overexpressed in a variety of different tumour types. METHODS: We have used a combination of RT-PCR, western blotting, tissue microarrays and metadata analysis of microarray data to study whether the FOXM1-PLK1 regulatory axis is upregulated and operational in oesophageal adenocarcinoma. RESULTS: FOXM1 and PLK1 are expressed in oesophageal adenocarcinoma-derived cell lines and demonstrate cross-regulatory interactions. Importantly, we also demonstrate the concomitant overexpression of FOXM1 and PLK1 in a large proportion of oesophageal adenocarcinoma samples. This co-association was extended to the additional FOXM1 target genes CCNB1, AURKB and CKS1. In a cohort of patients who subsequently underwent surgery, the expression of several FOXM1 target genes was prognostic for overall survival. CONCLUSIONS: FOXM1 and its target gene PLK1 are commonly overexpressed in oesophageal adenocarcinomas and this association can be extended to other FOXM1 target genes, providing potentially important biomarkers for predicting post-surgery disease survival.


Subject(s)
Adenocarcinoma/genetics , Cell Cycle Proteins/genetics , Esophageal Neoplasms/genetics , Forkhead Transcription Factors/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Adenocarcinoma/metabolism , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cohort Studies , Esophageal Neoplasms/metabolism , Forkhead Box Protein M1 , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Prognosis , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/metabolism , Up-Regulation , Polo-Like Kinase 1
5.
Br J Cancer ; 105(1): 124-30, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-21673681

ABSTRACT

BACKGROUND: Transcription factors often play important roles in tumourigenesis. Members of the PEA3 subfamily of ETS-domain transcription factors fulfil such a role and have been associated with tumour metastasis in several different cancers. Moreover, the activity of the PEA3 subfamily transcription factors is potentiated by Ras-ERK pathway signalling, which is itself often deregulated in tumour cells. METHODS: Immunohistochemical patterns of PEA3 expression and active ERK signalling were analysed and mRNA expression levels of PEA3, ER81, MMP-1 and MMP-7 were determined in gastric adenocarcinoma samples. RESULTS: Here, we have studied the expression of the PEA3 subfamily members PEA3/ETV4 and ER81/ETV1 in gastric adenocarcinomas. PEA3 is upregulated at the protein level in gastric adenocarcinomas and both PEA3/ETV4 and ER81/ETV1 are upregulated at the mRNA level in gastric adenocarcinoma tissues. This increased expression correlates with the expression of a target gene associated with metastasis, MMP-1. Enhanced ERK signalling is also more prevalent in late-stage gastric adenocarcinomas, and the co-association of ERK signalling and PEA3 expression also occurs in late-stage gastric adenocarcinomas. Furthermore, the co-association of ERK signalling and PEA3 expression correlates with decreased survival rates. CONCLUSIONS: This study shows that members of the PEA3 subfamily of transcription factors are upregulated in gastric adenocarcinomas and that the simultaneous upregulation of PEA3 expression and ERK pathway signalling is indicative of late-stage disease and a poor survival prognosis.


Subject(s)
DNA-Binding Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 7/metabolism , Stomach Neoplasms/metabolism , Transcription Factors/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Case-Control Studies , DNA-Binding Proteins/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Gastric Mucosa/metabolism , Humans , Immunoenzyme Techniques , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 7/genetics , Prognosis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcription Factors/genetics
6.
Biochem Soc Trans ; 35(Pt 6): 1414-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18031234

ABSTRACT

The modification of proteins by SUMO (small ubiquitin-related modifier) conjugation is becoming increasingly recognized as an important regulatory event. Protein SUMOylation can control a whole range of activities, including subcellular localization, protein-protein interactions and enzymatic activity. However, the SUMOylation process can itself be controlled. In the present review, the mechanisms through which protein SUMOylation is regulated are discussed, with particular emphasis on the impact of signalling pathways. A major point of regulation of the SUMO pathway is through targeting the E3 ligases, and a number of different ways to achieve this have been identified. More generally, the MAPK (mitogen-activated protein kinase) pathways represent one way through which SUMOylation of specific proteins is controlled, by using molecular mechanisms that at least in part also function by modifying the activity of SUMO E3 ligases. Further intricacies in signalling pathway interactions are hinted at through the growing number of examples of cross-talk between different post-translational modifications and SUMO modification.


Subject(s)
Signal Transduction/physiology , Small Ubiquitin-Related Modifier Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism
7.
FEBS Lett ; 581(6): 1233-42, 2007 Mar 20.
Article in English | MEDLINE | ID: mdl-17346708

ABSTRACT

GTF2IRD1 is a member of a family of transcription factors whose defining characteristic is varying numbers of a helix-loop-helix like motif, the I-repeat. Here, we present functional analysis of human GTF2IRD1 in regulation of three genes (HOXC8, GOOSECOID and TROPONIN I(SLOW)). We define a regulatory motif (GUCE-GTF2IRD1 Upstream Control Element) common to all three genes. GUCE is bound in vitro by domain I-4 of GTF2IRD1 and mediates transcriptional regulation by GTF2IRD1 in vivo. Definition of this site will assist in identification of other downstream targets of GTF2IRD1 and elucidation of its role in the human developmental disorder Williams-Beuren syndrome.


Subject(s)
Conserved Sequence , DNA/genetics , Gene Expression Regulation , Muscle Proteins/physiology , Nuclear Proteins/physiology , Trans-Activators/physiology , Transcription, Genetic , Goosecoid Protein/genetics , Homeodomain Proteins/genetics , Humans , Protein Binding , Regulatory Elements, Transcriptional , Troponin I/genetics , Williams Syndrome/genetics
8.
Article in English | MEDLINE | ID: mdl-16568956

ABSTRACT

The SUMO modification pathway has been linked with controlling the activity of numerous transcriptional regulatory proteins. In the majority of substrates studied so far, sumoylation imparts repressive properties. In several cases, part of this mechanism has been shown to be due to SUMO-dependent recruitment of histone deacetylases (HDACs). This is exemplified by the transcription factor Elk-1, where HDAC-2 is specifically recruited in response to sumoylation. Importantly, activation of the ERK MAP kinase pathway leads to Elk-1 desumoylation and HDAC loss. Furthermore, PIAS proteins can regulate the activities of transcription factors in SUMO-dependent and -independent manners. Further links between the MAP kinase pathways and PIAS proteins have been uncovered, suggesting a complex interplay been the MAP kinase and SUMO modification pathways. Here we discuss the current evidence suggesting links between the SUMO and MAP kinase pathways and point to other potential regulatory events and how these might be affected in cancer.


Subject(s)
MAP Kinase Signaling System/physiology , Regulatory Elements, Transcriptional/genetics , Repressor Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/metabolism , Repressor Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , ets-Domain Protein Elk-1/genetics , ets-Domain Protein Elk-1/metabolism
9.
Nat Rev Mol Cell Biol ; 2(11): 827-37, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11715049

ABSTRACT

ETS-domain transcription-factor networks represent a model for how combinatorial gene expression is achieved. These transcription factors interact with a multitude of co-regulatory partners to elicit gene-specific responses and drive distinct biological processes. These proteins are controlled by a complex series of inter and intramolecular interactions, and signalling pathways impinge on these proteins to further regulate their action.


Subject(s)
Proto-Oncogene Proteins/physiology , Transcription Factors/physiology , Animals , DNA/metabolism , Humans , Models, Biological , Models, Molecular , Multigene Family , Neoplasms/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-ets
10.
Mol Cell Biol ; 21(8): 2802-14, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11283259

ABSTRACT

The transcriptional status of eukaryotic genes is determined by a balance between activation and repression mechanisms. The nuclear hormone receptors represent classical examples of transcription factors that can regulate this balance by recruiting corepressor and coactivator complexes in a ligand-dependent manner. Here, we demonstrate that the equilibrium between activation and repression via a single transcription factor, Elk-1, is altered following activation of the Erk mitogen-activated protein kinase cascade. In addition to its C-terminal transcriptional activation domain, Elk-1 contains an N-terminal transcriptional repression domain that can recruit the mSin3A-histone deacetylase 1 corepressor complex. Recruitment of this corepressor is enhanced in response to activation of the Erk pathway in vivo, and this recruitment correlates kinetically with the shutoff of one of its target promoters, c-fos. Elk-1 therefore undergoes temporal activator-repressor switching and contributes to both the activation and repression of target genes following growth factor stimulation.


Subject(s)
DNA-Binding Proteins , Histone Deacetylases/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Biological Transport, Active , Cell Line , Epidermal Growth Factor/pharmacology , Histone Deacetylase 1 , Histone Deacetylases/chemistry , Histone Deacetylases/genetics , Humans , In Vitro Techniques , Kinetics , Macromolecular Substances , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Mitogen-Activated Protein Kinases/metabolism , Peptide Mapping , Promoter Regions, Genetic , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex , Trans-Activators/chemistry , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription, Genetic , ets-Domain Protein Elk-1
11.
Nature ; 409(6823): 1067-70, 2001 Feb 22.
Article in English | MEDLINE | ID: mdl-11234019

ABSTRACT

The p16INK4a cyclin-dependent kinase inhibitor is implicated in replicative senescence, the state of permanent growth arrest provoked by cumulative cell divisions or as a response to constitutive Ras-Raf-MEK signalling in somatic cells. Some contribution to senescence presumably underlies the importance of p16INK4a as a tumour suppressor but the mechanisms regulating its expression in these different contexts remain unknown. Here we demonstrate a role for the Ets1 and Ets2 transcription factors based on their ability to activate the p16INK4a promoter through an ETS-binding site and their patterns of expression during the lifespan of human diploid fibroblasts. The induction of p16INK4a by Ets2, which is abundant in young human diploid fibroblasts, is potentiated by signalling through the Ras-Raf-MEK kinase cascade and inhibited by a direct interaction with the helix-loop-helix protein Id1 (ref. 11). In senescent cells, where the Ets2 levels and MEK signalling decline, the marked increase in p16INK4a expression is consistent with the reciprocal reduction of Id1 and accumulation of Ets1.


Subject(s)
Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation , MAP Kinase Kinase Kinase 1 , Proto-Oncogene Proteins/physiology , Repressor Proteins , Trans-Activators/physiology , Transcription Factors/physiology , Animals , Cell Line , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Fibroblasts , Humans , Inhibitor of Differentiation Protein 1 , MAP Kinase Signaling System , Mice , Promoter Regions, Genetic , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Protein c-ets-2 , Proto-Oncogene Proteins c-ets , Proto-Oncogene Proteins c-raf/metabolism , ras Proteins/metabolism
12.
J Biol Chem ; 276(19): 16207-15, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11278941

ABSTRACT

The control of DNA binding by eukaryotic transcription factors represents an important regulatory mechanism. Many transcription factors are controlled by cis-acting autoinhibitory modules that are thought to act by blocking promiscuous DNA binding in the absence of appropriate regulatory cues. Here, we have investigated the determinants and regulation of the autoinhibitory mechanism employed by the ETS-domain transcription factor, PEA3. DNA binding is inhibited by a module composed of a combination of two short motifs located on either side of the ETS DNA-binding domain. A second type of protein, Ids, can act in trans to mimic the effect of these cis-acting inhibitory motifs and reduce DNA binding by PEA3. By using a one-hybrid screen, we identified the basic helix-loop-helix-leucine zipper transcription factor USF-1 as an interaction partner for PEA3. PEA3 and USF-1 form DNA complexes in a cooperative manner. Moreover, the formation of ternary PEA3.USF-1.DNA complexes requires parts of the same motifs in PEA3 that form the autoinhibitory module. Thus the binding of USF-1 to PEA3 acts as a switch that modifies the autoinhibitory motifs in PEA3 to first relieve their inhibitory action, and second, promote ternary nucleoprotein complex assembly.


Subject(s)
DNA-Binding Proteins , DNA/metabolism , Oncogene Proteins , Transcription Factors/chemistry , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Cloning, Molecular , DNA/chemistry , Escherichia coli/genetics , Genes, Reporter , Helix-Loop-Helix Motifs , Humans , Leucine Zippers , Mice , Molecular Sequence Data , Plasmids , Proto-Oncogene Proteins c-ets , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Transcription Factors/genetics , Transfection , Upstream Stimulatory Factors
13.
Mol Cell Biol ; 21(2): 524-33, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11134340

ABSTRACT

The Id subfamily of helix-loop-helix (HLH) proteins plays a fundamental role in the regulation of cellular proliferation and differentiation. The major mechanism by which Id proteins are thought to inhibit differentiation is through interaction with other HLH proteins and inhibition of their DNA-binding activity. However, Id proteins have also been shown to interact with other proteins involved in regulating cellular proliferation and differentiation, suggesting a more widespread regulatory function. In this study we demonstrate functional interactions between Id proteins and members of the Pax-2/-5/-8 subfamily of paired-domain transcription factors. Members of the Pax transcription factor family have key functions in regulating several developmental processes exemplified by B lymphopoiesis, in which Pax-5 plays an essential role. Id proteins bind to Pax proteins in vitro and in vivo. Binding occurs through the paired DNA-binding domain of the Pax proteins and results in the disruption of DNA-bound complexes containing Pax-2, Pax-5, and Pax-8. In vivo, Id proteins modulate the transcriptional activity mediated by Pax-5 complexes on the B-cell-specific mb-1 promoter. Our results therefore demonstrate a novel facet of Id function in regulating cellular differentiation by functionally antagonizing the action of members of the Pax transcription factor family.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Helix-Loop-Helix Motifs , Neoplasm Proteins , Nuclear Proteins/antagonists & inhibitors , Repressor Proteins , Trans-Activators/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , 3T3 Cells , Animals , Antigens, CD/genetics , Base Sequence , CD79 Antigens , COS Cells , DNA/genetics , DNA/metabolism , Inhibitor of Differentiation Protein 1 , Inhibitor of Differentiation Protein 2 , Inhibitor of Differentiation Proteins , Mice , Nuclear Proteins/metabolism , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/metabolism , PAX2 Transcription Factor , PAX5 Transcription Factor , PAX8 Transcription Factor , Paired Box Transcription Factors , Precipitin Tests , Promoter Regions, Genetic/genetics , Protein Binding , Proto-Oncogene Proteins/metabolism , Receptors, Antigen, B-Cell/genetics , Trans-Activators/metabolism , ets-Domain Protein Elk-1
14.
J Biol Chem ; 276(2): 965-73, 2001 Jan 12.
Article in English | MEDLINE | ID: mdl-11029469

ABSTRACT

MAPK pathways play important roles in regulating the key cellular processes of proliferation, differentiation, and apoptosis. There are multiple MAPK pathways, which are subject to different regulatory cues. It is important that these pathways maintain specificity in signaling to elicit the activation of a specific program of gene expression. MAPK-docking domains in several transcription factors have been shown to play important roles in determining the specificity and efficiency of their phosphorylation by MAPKs. Here we investigate the mechanisms by which MAPKs are targeted to the ETS domain transcription factor SAP-1. We demonstrate that SAP-1 contains two different domains that are required for its efficient phosphorylation in vitro and activation in vivo by ERK2 and a subset of p38 MAPKs. The D-domain is closely related to other MAPK-docking domains, but exhibits a novel specificity and serves to promote selective targeting of ERK2, p38alpha, and p38beta(2) to SAP-1. A second important region, the FXF motif, also plays an important role in directing MAPKs to phosphorylate SAP-1. The FXF motif promotes targeting by ERK2 and, to a lesser extent, p38alpha, but not p38beta(2). Our data therefore demonstrate that a modular system of motifs is responsible for directing specific MAPK subtypes to SAP-1, but also point to important distinctions in the mechanism of action of the D-domain and FXF motif.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Chlorocebus aethiops , Genes, Reporter , Kinetics , MAP Kinase Kinase 6 , Mitogen-Activated Protein Kinase 1/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Substrate Specificity , Transcription Factors/chemistry , Transfection , ets-Domain Protein Elk-4 , p38 Mitogen-Activated Protein Kinases
16.
Trends Biochem Sci ; 25(9): 448-53, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10973059

ABSTRACT

Signalling specificity in eukaryotic cells is maintained by several mechanisms. One mechanism by which mitogen-activated protein (MAP) kinases ensure their specificity of action is by interacting with their substrates through docking domains. These docking domains recruit the kinases to the correct substrates and enhance their fidelity and efficiency of action. Additional specificity determinants in the substrates serve to enhance the specificity of substrate phosphorylation by MAP kinases further.


Subject(s)
MAP Kinase Signaling System , Amino Acid Sequence , Animals , Evolution, Molecular , Molecular Sequence Data , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Signal Transduction , Substrate Specificity , Transcription Factors/chemistry , Transcription Factors/metabolism
17.
EMBO J ; 19(14): 3750-61, 2000 Jul 17.
Article in English | MEDLINE | ID: mdl-10899128

ABSTRACT

In the yeast Saccharomyces cerevisiae, the MADS-box protein Mcm1, which is highly related to mammalian SRF (serum response factor), forms a ternary complex with SFF (Swi five factor) to regulate the cell cycle expression of genes such as SWI5, CLB2 and ACE2. Here we show that the forkhead protein Fkh2 is a component of SFF and is essential for ternary complex formation on the SWI5 and ACE2 promoters. Fkh2 is essential for the correct cell cycle periodicity of SWI5 and CLB2 gene expression and is phosphorylated with a timing that is consistent with a role in this expression. Furthermore, investigation of the relationship between Fkh2 and a related forkhead protein Fkh1 demonstrates that these proteins act in overlapping pathways to regulate cell morphology and cell separation. This is the first example of a eukaryotic transcription factor complex containing both a MADS-box and a forkhead protein, and it has important implications for the regulation of mammalian gene expression.


Subject(s)
Cell Cycle Proteins , Cell Cycle/genetics , Gene Expression Regulation, Fungal , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Transcription Factors/chemistry , Transcription Factors/metabolism , Cell Nucleus/metabolism , Consensus Sequence/genetics , Cyclin B/genetics , Cyclin B/metabolism , Cyclins/genetics , DNA, Fungal/genetics , DNA, Fungal/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Forkhead Transcription Factors , Fungal Proteins/genetics , Fungal Proteins/metabolism , G2 Phase/genetics , Gene Deletion , Genes, Fungal/genetics , Minichromosome Maintenance 1 Protein , Nuclear Proteins/genetics , Phosphorylation , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Messenger/metabolism , Response Elements/genetics , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Spindle Apparatus/metabolism , Transcription Factors/genetics
18.
EMBO J ; 19(11): 2615-28, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10835359

ABSTRACT

The solution structure of the 33 kDa complex between the dimeric DNA-binding core domain of the transcription factor MEF2A (residues 1-85) and a 20mer DNA oligonucleotide comprising the consensus sequence CTA(A/T)(4)TAG has been solved by NMR. The protein comprises two domains: a MADS-box (residues 1-58) and a MEF2S domain (residues 59-73). Recognition and specificity are achieved by interactions between the MADS-box and both the major and minor grooves of the DNA. A number of critical differences in protein-DNA contacts observed in the MEF2A-DNA complex and the DNA complexes of the related MADS-box transcription factors SRF and MCM1 provide a molecular explanation for modulation of sequence specificity and extent of DNA bending ( approximately 15 versus approximately 70 degrees ). The structure of the MEF2S domain is entirely different from that of the equivalent SAM domain in SRF and MCM1, accounting for the absence of cross-reactivity with other proteins that interact with these transcription factors.


Subject(s)
DNA-Binding Proteins/chemistry , DNA/chemistry , Transcription Factors/chemistry , Amino Acid Sequence , Cross Reactions , DNA/metabolism , DNA-Binding Proteins/metabolism , Dimerization , Humans , MADS Domain Proteins , MEF2 Transcription Factors , Macromolecular Substances , Magnetic Resonance Spectroscopy , Minichromosome Maintenance 1 Protein , Models, Molecular , Molecular Sequence Data , Myogenic Regulatory Factors , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nucleic Acid Conformation/drug effects , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Regulatory Sequences, Nucleic Acid , Sequence Alignment , Sequence Homology, Amino Acid , Serum Response Factor , Solutions , Structure-Activity Relationship , Transcription Factors/classification , Transcription Factors/metabolism , Transcription Factors/pharmacology
19.
Mech Dev ; 90(2): 237-52, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10640707

ABSTRACT

Fli-1 is an ETS-domain transcription factor whose locus is disrupted in Ewing's Sarcoma and F-MuLV induced erythroleukaemia. To gain a better understanding of its normal function, we have isolated the zebrafish homologue. Similarities with other vertebrates, in the amino acid sequence and DNA binding properties of Fli-1 from zebrafish, suggest that its function has been conserved during vertebrate evolution. The initial expression of zebrafish fli-1 in the posterior lateral mesoderm overlaps with that of gata2 in a potential haemangioblast population which likely contains precursors of blood and endothelium. Subsequently, fli-1 and gata2 expression patterns diverge, with separate fli-1 and gata2 expression domains arising in the developing vasculature and in sites of blood formation respectively. Elsewhere in the embryo, fli-1 is expressed in sites of vasculogenesis. The expression of fli-1 was investigated in a number of zebrafish mutants, which affect the circulatory system. In cloche, endothelium is absent and blood is drastically reduced. In contrast to the blood and endothelial markers that have been studied previously, fli-1 expression was initiated normally in cloche embryos, indicating that induction of fli-1 is one of the earliest indicators of haemangioblast formation. Furthermore, although fli-1 expression in the trunk was not maintained, the normal expression pattern in the anterior half of the embryo was retained. These anterior cells did not, however, condense to form blood vessels. These data indicate that cloche has previously unsuspected roles at multiple stages in the formation of the vasculature. Analysis of fli-1 expression in midline patterning mutants floating head and squint, confirms a requirement for the notochord in the formation of the dorsal-aorta. The formation of endothelium in one-eyed pinhead, cyclops and squint embryos indicates a novel role for the endoderm in the formation of the axial vein. The phenotype of sonic-you mutants implies a likely role for Sonic Hedgehog in mediating these processes.


Subject(s)
DNA-Binding Proteins/genetics , Neovascularization, Physiologic/physiology , Proto-Oncogene Proteins , Trans-Activators/genetics , Zebrafish Proteins , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , GATA2 Transcription Factor , Gene Expression , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins , Mice , Molecular Sequence Data , Mutation , Nodal Signaling Ligands , Proto-Oncogene Protein c-fli-1 , Sequence Homology, Amino Acid , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Zebrafish
20.
Nucleic Acids Res ; 28(2): 560-9, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10606656

ABSTRACT

The mechanisms of multicomponent transcription factor complex assembly are currently poorly defined. A paradigm for this type of complex is the ETS-domain transcription factor Elk-1 and the MADS-box transcription factor SRF which form a ternary complex with the c- fos serum response element (SRE). In this study we have analysed how a different ETS-domain transcription factor Fli-1 interacts with SRF to form ternary complexes with this element. Two regions of Fli-1 that are required for ternary complex formation have been identified. These SRF binding motifs are located on either side of the ETS DNA-binding domain. Hydrophobic amino acids within these motifs have been identified that play important roles in binding to SRF and ternary complex formation. By using Fli-1 derivatives with mutations in the N-terminal SRF binding motif, the significance of Fli-1-SRF interactions in recruitment of Fli-1 to the c- fos SRE in vivo has been demonstrated. Collectively our data provide a model of how Fli-1 interacts with SRF that differs significantly from the mechanism used by a different ETS-domain protein, Elk-1.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins , Trans-Activators/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Binding Sites , DNA-Binding Proteins/genetics , Humans , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins/genetics , Protein Binding , Proto-Oncogene Protein c-fli-1 , Sequence Homology, Amino Acid , Serum Response Factor
SELECTION OF CITATIONS
SEARCH DETAIL
...