Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Dokl Biochem Biophys ; 482(1): 255-260, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30397887

ABSTRACT

It is shown that mutations in two GC-rich sequences (GC-boxes) from the promoter region of the yellow gene during enhancer-dependent transcription activation do not affect the basal level of the yellow gene transcription but destabilize the interaction between the enhancers and the promoter.


Subject(s)
Enhancer Elements, Genetic , GC Rich Sequence/physiology , Promoter Regions, Genetic , Animals , Drosophila melanogaster/genetics , TATA Box/genetics , Transcription, Genetic/drug effects
2.
Nucleic Acids Res ; 42(7): 4527-45, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24470146

ABSTRACT

Most RNA viruses infecting mammals and other vertebrates show profound suppression of CpG and UpA dinucleotide frequencies. To investigate this functionally, mutants of the picornavirus, echovirus 7 (E7), were constructed with altered CpG and UpA compositions in two 1.1-1.3 Kbase regions. Those with increased frequencies of CpG and UpA showed impaired replication kinetics and higher RNA/infectivity ratios compared with wild-type virus. Remarkably, mutants with CpGs and UpAs removed showed enhanced replication, larger plaques and rapidly outcompeted wild-type virus on co-infections. Luciferase-expressing E7 sub-genomic replicons with CpGs and UpAs removed from the reporter gene showed 100-fold greater luminescence. E7 and mutants were equivalently sensitive to exogenously added interferon-ß, showed no evidence for differential recognition by ADAR1 or pattern recognition receptors RIG-I, MDA5 or PKR. However, kinase inhibitors roscovitine and C16 partially or entirely reversed the attenuated phenotype of high CpG and UpA mutants, potentially through inhibition of currently uncharacterized pattern recognition receptors that respond to RNA composition. Generating viruses with enhanced replication kinetics has applications in vaccine production and reporter gene construction. More fundamentally, the findings introduce a new evolutionary paradigm where dinucleotide composition of viral genomes is subjected to selection pressures independently of coding capacity and profoundly influences host-pathogen interactions.


Subject(s)
Dinucleoside Phosphates/physiology , Enterovirus B, Human/physiology , GC Rich Sequence/physiology , RNA, Viral/chemistry , Virus Replication , Base Composition , Cell Line , Enterovirus B, Human/genetics , Mutation
3.
PLoS One ; 8(8): e73121, 2013.
Article in English | MEDLINE | ID: mdl-24015290

ABSTRACT

G-quadruplexes are tetraplex structures of nucleic acids that can form in G-rich sequences. Their presence and functional role have been established in telomeres, oncogene promoters and coding regions of the human chromosome. In particular, they have been proposed to be directly involved in gene regulation at the level of transcription. Because the HIV-1 Nef protein is a fundamental factor for efficient viral replication, infectivity and pathogenesis in vitro and in vivo, we investigated G-quadruplex formation in the HIV-1 nef gene to assess the potential for viral inhibition through G-quadruplex stabilization. A comprehensive computational analysis of the nef coding region of available strains showed the presence of three conserved sequences that were uniquely clustered. Biophysical testing proved that G-quadruplex conformations were efficiently stabilized or induced by G-quadruplex ligands in all three sequences. Upon incubation with a G-quadruplex ligand, Nef expression was reduced in a reporter gene assay and Nef-dependent enhancement of HIV-1 infectivity was significantly repressed in an antiviral assay. These data constitute the first evidence of the possibility to regulate HIV-1 gene expression and infectivity through G-quadruplex targeting and therefore open a new avenue for viral treatment.


Subject(s)
GC Rich Sequence/physiology , Gene Expression Regulation, Viral/physiology , HIV-1/physiology , Virus Replication/physiology , nef Gene Products, Human Immunodeficiency Virus/metabolism , HEK293 Cells , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/therapy , HIV-1/chemistry , Humans , nef Gene Products, Human Immunodeficiency Virus/chemistry , nef Gene Products, Human Immunodeficiency Virus/genetics
4.
Genome Res ; 22(1): 51-63, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22006216

ABSTRACT

Analysis of diverse eukaryotes has revealed that recombination events cluster in discrete genomic locations known as hotspots. In humans, a zinc-finger protein, PRDM9, is believed to initiate recombination in >40% of hotspots by binding to a specific DNA sequence motif. However, the PRDM9 coding sequence is disrupted in the dog genome assembly, raising questions regarding the nature and control of recombination in dogs. By analyzing the sequences of PRDM9 orthologs in a number of dog breeds and several carnivores, we show here that this gene was inactivated early in canid evolution. We next use patterns of linkage disequilibrium using more than 170,000 SNP markers typed in almost 500 dogs to estimate the recombination rates in the dog genome using a coalescent-based approach. Broad-scale recombination rates show good correspondence with an existing linkage-based map. Significant variation in recombination rate is observed on the fine scale, and we are able to detect over 4000 recombination hotspots with high confidence. In contrast to human hotspots, 40% of canine hotspots are characterized by a distinct peak in GC content. A comparative genomic analysis indicates that these peaks are present also as weaker peaks in the panda, suggesting that the hotspots have been continually reinforced by accelerated and strongly GC biased nucleotide substitutions, consistent with the long-term action of biased gene conversion on the dog lineage. These results are consistent with the loss of PRDM9 in canids, resulting in a greater evolutionary stability of recombination hotspots. The genetic determinants of recombination hotspots in the dog genome may thus reflect a fundamental process of relevance to diverse animal species.


Subject(s)
Evolution, Molecular , Genome/physiology , Genomic Instability/physiology , Histone-Lysine N-Methyltransferase/genetics , Polymorphism, Single Nucleotide , Recombination, Genetic/physiology , Animals , Dogs , GC Rich Sequence/physiology , Humans
5.
Biometals ; 23(6): 1075-84, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20577783

ABSTRACT

The DNA binding and cleavage properties of quercetin nickel (II) complex have been studied, but little attention has been devoted to the relationship between antitumor activity of this complex and DNA-binding properties. In the present study, we report that quercetin nickel (II) complex showed significant cytotoxicity against three tumor cell lines (HepG2, SMMC7721 and A549). Hoechst33258 and AO/EB staining showed HepG2 cells underwent the typical morphologic changes of apoptosis characterized by nuclear shrinkage, chromatin condensation, or fragmentation after exposure to quercetin nickel (II) complex. We also demonstrate that the levels of survivin and bcl-2 protein expression in HepG2 cells decreased concurrently, and the levels of p53 protein increased significantly after treatment with quercetin nickel (II) complex by immunocytochemistry analysis. The relative activity of caspase-3 and caspase-9 increased significantly after treatment with the complex. Furthermore, fluorescence measurements and molecular modeling were performed to learn that the complex could be preferentially bound to DNA in GC region. These results imply that quercetin nickel (II) complex may intercalate into the GC-rich core promoter region of survivin, down-regulating survivin gene expression and promoting tumor cells apoptosis. So our results suggest that antitumor activity of quercetin nickel (II) complex might be related to its intercalation into DNA and DNA-binding selectivity, and that the complex may be a promising agent for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/therapeutic use , Microtubule-Associated Proteins/genetics , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Quercetin/analogs & derivatives , Apoptosis/drug effects , Cell Line, Tumor , Coordination Complexes/pharmacology , DNA, Neoplasm/metabolism , Down-Regulation , GC Rich Sequence/physiology , Hep G2 Cells/drug effects , Humans , Inhibitor of Apoptosis Proteins , Organometallic Compounds/therapeutic use , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Quercetin/chemistry , Quercetin/pharmacology , Quercetin/therapeutic use , Survivin
6.
Exp Parasitol ; 124(3): 346-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19931528

ABSTRACT

Recent findings associate transcription start in trypanosomatids with chromatin regions containing modified and variant histones. TATA-binding protein (TBP) and other fundamental transcription factors have been also found at these Transcription Start Sites (TSS). Results of Systematic Evolution of Ligands by Exponential Enrichment (SELEX) experiments show that Trypanosoma cruzi TBP (TcTBP) has an in vitro binding preference for G-rich sequences. This finding correlates with the presence of G-rich stretches at the Strand Switch Regions (SSR) and at some putative internal TSS in Trypanosoma brucei and Leishmania. A scanning study of partially assembled T. cruzi genomic contigs determined the presence of G-rich stretches in the coding strands. TcTBP affinity for G-rich sequences suggests that this factor could play a role in locating the initiation complex in the right TSS, probably by "sensing" the G-content on the strand to be transcribed.


Subject(s)
GC Rich Sequence/physiology , TATA-Box Binding Protein/metabolism , Trypanosoma cruzi/metabolism , Consensus Sequence , Trypanosoma cruzi/genetics
7.
Gene ; 407(1-2): 54-62, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-17964742

ABSTRACT

SSRs (simple sequence repeats) have been shown to have a variety of effects on an organism. In this study, we compared SSRs in housekeeping and tissue-specific genes in human and mouse, in terms of SSR types and distributions in different regions including 5'-UTRs, introns, coding exons, 3'-UTRs, and upstream regions. Among all these regions, SSRs in the 5'-UTR show the most distinction between housekeeping genes and tissue-specific genes in both densities and repeat types. Specifically, SSR densities in 5'-UTRs in housekeeping genes are about 1.7 times higher than those in tissue-specific genes, in contrast to the 0.8-1.2 times differences between the two classes of genes in other regions. Tri-SSRs in 5'-UTRs of housekeeping genes are more GC rich than those of tissue-specific genes and CGG, the dominant type of tri-SSR in 5'-UTR, accounts for 74-79% of the tri-SSRs in housekeeping genes, as compared to 42-57% in tissue-specific genes. 75% of the tri-SSRs in the 5'-UTR of housekeeping genes have 4-5 repeat units, versus the 86-90% in tissue-specific genes. Taken together, our results suggest that SSRs may have an effect on gene expression and may play an important role in contributing to the different expression profiles between housekeeping and tissue-specific genes.


Subject(s)
5' Untranslated Regions/chemistry , GC Rich Sequence/physiology , Gene Expression Regulation , Microsatellite Repeats/physiology , Animals , Base Sequence , GC Rich Sequence/genetics , Genes , Humans , Mice
8.
J Biol Chem ; 282(22): 15981-94, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17426024

ABSTRACT

Manganese superoxide dismutase (MnSOD), a mitochondrial antioxidant enzyme, is necessary for survival of aerobic life. Previously, we demonstrated that a Sp1-based promoter is essential for constitutive transcription and a NF-kappaB-based intronic enhancer is responsible for cytokine-mediated induction. Here we show that nucleophosmin (NPM), a RNA-binding protein, binds to an 11G single-stranded loop in the promoter region and serves to integrate the Sp1 and NF-kappaB responses. Disruption of the loop structure causes a reduction of both constitutive and inductive transcription due to loss of the binding motif for NPM. Interaction of NF-kappaB.NPM.Sp1 facilitated by binding of NPM to the loop structure in the promoter region appears to comprise the basic complex for the transcriptional stimulation. These results suggest a novel molecular mechanism for communication between the enhancer and the GC-rich promoter.


Subject(s)
DNA, Single-Stranded/genetics , Enhancer Elements, Genetic/physiology , Nucleic Acid Conformation , Response Elements/physiology , Superoxide Dismutase/genetics , Transcription, Genetic/physiology , Cell Line, Transformed , DNA, Single-Stranded/metabolism , GC Rich Sequence/physiology , Humans , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Nucleophosmin , Protein Binding/physiology , Sp1 Transcription Factor/metabolism , Superoxide Dismutase/biosynthesis
9.
Endocrinology ; 147(2): 899-911, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16254027

ABSTRACT

Establishment of pregnancy in ruminants results from paracrine signaling by interferon tau (IFNT) from the conceptus to uterine endometrial luminal epithelia (LE) that prevents release of luteolytic prostaglandin F(2alpha) pulses. In cyclic and pregnant ewes, progesterone down-regulates progesterone receptor (PGR) gene expression in LE. In cyclic ewes, loss of PGR allows for increases in estrogen receptor alpha (ESR1) and then oxytocin receptor (OXTR) gene expression followed by oxytocin-induced prostaglandin F(2alpha) pulses. In pregnant ewes, IFNT inhibits transcription of the ESR1 gene, which presumably inhibits OXTR gene transcription. Alternatively, IFNT may directly inhibit OXTR gene transcription. The 5' promoter/enhancer region of the ovine OXTR gene was cloned and found to contain predicted binding sites for activator protein 1, SP1, and PGR, but not for ESR1. Deletion analysis showed that the basal promoter activity was dependent on the region from -144 to -4 bp that contained only SP1 sites. IFNT did not affect activity of the OXTR promoter. In cells transfected with ESR1, E2, and ICI 182,780 increased promoter activity due to GC-rich SP1 binding sites at positions -104 and -64. Mutation analyses showed that the proximal SP1 sites mediated ESR1 action as well as basal activity of the promoter. In response to progesterone, progesterone receptor B also increased OXTR promoter activity. SP1 protein was constitutively expressed and abundant in the LE of the ovine uterus. These results support the hypothesis that the antiluteolytic effects of IFNT are mediated by direct inhibition or silencing of ESR1 gene transcription, thereby precluding ESR1/SP1 from stimulating OXTR gene transcription.


Subject(s)
Estrogen Receptor alpha/metabolism , Pregnancy, Animal/physiology , Promoter Regions, Genetic/physiology , Receptors, Oxytocin/metabolism , Sheep/physiology , Sp1 Transcription Factor/metabolism , Animals , Estrogens/physiology , Estrous Cycle/physiology , Feedback, Physiological/physiology , Female , GC Rich Sequence/physiology , Gene Silencing/physiology , Interferon Type I/physiology , Male , Pregnancy , Pregnancy Proteins/physiology , Progesterone/physiology , Receptors, Oxytocin/genetics , Transcription Factor AP-1/metabolism , Transcriptional Activation/physiology
10.
Bioorg Med Chem Lett ; 15(12): 3151-6, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15876536

ABSTRACT

Hairpin conjugates of achiral seco-cyclopropaneindoline-2-benzofurancarboxamide (achiral seco-CI-Bf) and three diamides (ImPy 1, PyIm 2, and PyPy 3, where Py is pyrrole, and Im is imidazole), linked by a gamma-aminobutyrate group, were synthesized. The sequence-specific covalent alkylation of the achiral CI moiety with adenine-N3 in the minor groove was ascertained by thermally induced DNA cleavage experiments. The results provide evidence that hairpin conjugates of achiral seco-CI-Bf-gamma-polyamides could be tailored to target specific DNA sequences according to a set of general rules: the achiral CI moiety selectively reacts with adenine-N3, a stacked pair of imidazole/benzofuran prefers a G/C base pair, and a pyrrole/benzofuran prefers an A/T or T/A base pair. Models for the binding of hairpin conjugates 1-3 with sequences 5'-TCA(888)G-3', 5'-CAA(857)C-3', and 5'-TTA(843)C-3' are proposed.


Subject(s)
Amides/metabolism , Benzofurans/metabolism , DNA/genetics , DNA/metabolism , Imidazoles/chemistry , Pyrroles/chemistry , AT Rich Sequence/physiology , Amides/chemistry , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Benzofurans/chemistry , Binding Sites , DNA/radiation effects , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , GC Rich Sequence/physiology , Hot Temperature , Molecular Conformation , Nucleic Acid Conformation , Promoter Regions, Genetic/genetics , Substrate Specificity , Thermodynamics
11.
Mol Immunol ; 41(9): 855-61, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15261457

ABSTRACT

Interferon regulatory factor-4 (IRF-4) is a lymphoid-specific transcription factor that plays crucial roles in the development and the functions of immune cells. B lymphocytes express IRF-4 constitutively. In this report, we investigated the transcriptional control of IRF-4 in B lymphocytes. Successive deletions of the IRF-4 promoter from -4799 revealed that the region between -51 and -28 (5'-CGCCCGCCCCAGGCCCCGCCCCA-3') was required for the basal promoter activity. Mutations in the distal and proximal sites of this GC-rich sequence resulted in 62 and 81% reductions in the IRF-4 promoter activity, respectively. EMSA observations revealed the formation of a protein complex with the corresponding DNA, which was sensitive to mutations in the GC-rich sequences. UV photocrosslinking assays identified a novel 60 kDa protein with a similar sequence preference. The possible involvement of this factor in the regulation of IRF-4 gene expression is discussed.


Subject(s)
B-Lymphocytes/metabolism , DNA-Binding Proteins/genetics , DNA/metabolism , Promoter Regions, Genetic , Transcription Factors/genetics , DNA-Binding Proteins/metabolism , GC Rich Sequence/genetics , GC Rich Sequence/physiology , Humans , Interferon Regulatory Factors , Transcription Factors/metabolism
12.
EMBO J ; 23(3): 541-51, 2004 Feb 11.
Article in English | MEDLINE | ID: mdl-14739937

ABSTRACT

Notch and bone morphogenetic protein signaling pathways are important for cellular differentiation, and both have been implicated in vascular development. In many cases the two pathways act similarly, but antagonistic effects have also been reported. The underlying mechanisms and whether this is caused by an interplay between Notch and BMP signaling is unknown. Here we report that expression of the Notch target gene, Herp2, is synergistically induced upon activation of Notch and BMP receptor signaling pathways in endothelial cells. The synergy is mediated via RBP-Jkappa/CBF-1 and GC-rich palindromic sites in the Herp2 promoter, as well as via interactions between the Notch intracellular domain and Smad that are stabilized by p/CAF. Activated Notch and its downstream effector Herp2 were found to inhibit endothelial cell (EC) migration. In contrast, BMP via upregulation of Id1 expression has been reported to promote EC migration. Interestingly, Herp2 was found to antagonize BMP receptor/Id1-induced migration by inhibiting Id1 expression. Our results support the notion that Herp2 functions as a critical switch downstream of Notch and BMP receptor signaling pathways in ECs.


Subject(s)
Endothelial Cells/physiology , Membrane Proteins/metabolism , Receptors, Growth Factor/metabolism , Signal Transduction/physiology , Animals , Bone Morphogenetic Protein Receptors , COS Cells , Cell Movement/genetics , Cell Movement/physiology , Chlorocebus aethiops , DNA-Binding Proteins/metabolism , Down-Regulation/physiology , GC Rich Sequence/physiology , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein , Inhibitor of Differentiation Protein 1 , Membrane Proteins/genetics , Mice , Nuclear Proteins/metabolism , Promoter Regions, Genetic/physiology , Receptors, Growth Factor/genetics , Receptors, Notch , Repressor Proteins/metabolism , Signal Transduction/genetics , Smad Proteins , Trans-Activators/metabolism , Transcription Factors/metabolism , Up-Regulation/physiology
13.
Blood ; 101(11): 4492-9, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12576331

ABSTRACT

Rearrangement of T-cell receptor (TCR) and immunoglobulin genes by a common V(D)J recombination machinery is regulated by developmentally specific chromatin changes at the target locus, a process associated with transcription. At the TCRbeta locus, the Ebeta enhancer and the Dbeta1 promoter regulate germline transcription originating near the TCR Dbeta1 gene segment. The Dbeta1 promoter contains 3 GC-rich motifs that bind a common set of nuclear proteins from pro-T-cell lines. Mutations that diminish the binding of nuclear proteins also diminish the activity of the Dbeta1 promoter in transcriptional reporter assays. Using a yeast one-hybrid approach, 3 Krüppel-like factors-KLF3, KLF5, and KLF6-and a novel zinc finger protein were identified in a thymus library, all of which bound the GC-rich motif in a sequence-specific manner. Of these genes, KLF5 mRNA was expressed in a restricted manner in lymphoid cells and tissues, with highest expression in pro-T-cell lines and Rag-deficient thymocytes. Antibody supershift studies and chromatin immunoprecipitation assay confirmed that KLF5 bound the Dbeta1 promoter. In reporter gene assays, KLF5 but not KLF6 efficiently transactivated the Dbeta1 promoter, whereas a dominant-negative KLF5 construct inhibited reporter expression. These data suggest that reiterated GC motifs contribute to germline TCRbeta transcription through binding of KLF5 and other Krüppel family members and that restricted expression of KLF5 may contribute to lineage-specific regulation of germline TCRbeta transcription.


Subject(s)
GC Rich Sequence/physiology , Genes, T-Cell Receptor beta , Promoter Regions, Genetic , Trans-Activators/physiology , Transcriptional Activation , Animals , DNA-Binding Proteins/metabolism , Kruppel-Like Transcription Factors , Mice , POU Domain Factors , Protein Binding , T-Lymphocytes/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription, Genetic , Tumor Cells, Cultured
14.
Am J Physiol Renal Physiol ; 284(1): F82-94, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12473536

ABSTRACT

We investigated the expression pattern and underlying mechanism that controls hepatocyte growth factor (HGF) receptor (c-met) expression in normal kidney and a variety of kidney cells. Immunohistochemical staining showed widespread expression of c-met in mouse kidney, a pattern closely correlated with renal expression of Sp1 and Sp3 transcription factors. In vitro, all types of kidney cells tested expressed different levels of c-met, which was tightly proportional to the cellular abundances of Sp1 and Sp3. Both Sp1 and Sp3 bound to the multiple GC boxes in the promoter region of the c-met gene. Coimmunoprecipitation suggested a physical interaction between Sp1 and Sp3. Functionally, Sp1 markedly stimulated c-met promoter activity. Although Sp3 only weakly activated the c-met promoter, its combination with Sp1 synergistically stimulated c-met transcription. Conversely, deprivation of Sp proteins by transfection of decoy Sp1 oligonucleotide or blockade of Sp1 binding with mithramycin A inhibited c-met expression. The c-met receptor in all types of kidney cells was functional and induced protein kinase B/Akt phosphorylation in a distinctly dynamic pattern after HGF stimulation. These results indicate that members of the Sp family of transcription factors play an important role in regulating constitutive expression of the c-met gene in all types of renal cells. Our findings suggest that HGF may have a broader spectrum of target cells and possess wider implications in kidney structure and function than originally thought.


Subject(s)
DNA-Binding Proteins/metabolism , Kidney/cytology , Kidney/physiology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-met/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Drosophila , Drosophila Proteins , Fibroblasts/cytology , Fibroblasts/physiology , GC Rich Sequence/physiology , Gene Expression/physiology , Glomerular Mesangium/cytology , Glomerular Mesangium/physiology , Humans , In Vitro Techniques , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/physiology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/physiology , Male , Mice , Mice, Inbred Strains , Oligonucleotides/pharmacology , Phosphorylation , Promoter Regions, Genetic/physiology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-met/metabolism , Rats , Sp3 Transcription Factor , Transcription, Genetic/physiology
15.
J Clin Endocrinol Metab ; 87(12): 5675-85, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12466371

ABSTRACT

G(s)alpha is the G protein subunit that stimulates adenylyl cyclase activity in the myometrium during pregnancy, raising intracellular levels of the smooth muscle relaxant cAMP. The promoter region of the gene encoding G(s)alpha is GC rich and contains multiple putative binding sites for the specificity protein (Sp) transcription factor family. In electrophoretic mobility shift assays, four of these Sp sites were bound by recombinant Sp1 protein. Binding was dependent on phosphorylation of Sp1 by protein kinase A. Phosphorylated Sp1-4 proteins were observed in extracts of cultured human myometrial cells, but in electrophoretic mobility shift assays G(s)alpha promoter sequence binding by Sp1 was not apparent. Instead, these assays showed phosphorylation-dependent G(s)alpha promoter binding by lower molecular weight myometrial proteins that could not be supershifted by antibodies specific to Sp1-4 proteins. To investigate the regulation of G(s)alpha expression, the GC-rich promoter region was used to direct transcription of a firefly luciferase reporter gene in transient transfection assays of primary human myometrial cell cultures, COS-7 and HEK 293 cells. Reporter gene expression was found to follow a biphasic response to forskolin and 8-bromo-cAMP, with an initial, concentration-dependent increase in luciferase activity, followed by a prolonged decrease. In myometrial cells, this pattern was also seen in response to treatment with human chorionic gonadotropin.


Subject(s)
Adenylyl Cyclases/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Labor, Obstetric/metabolism , Myometrium/metabolism , Pregnancy/metabolism , Promoter Regions, Genetic/physiology , Transcription, Genetic/physiology , Animals , Base Sequence/genetics , COS Cells , Cells, Cultured , Cyclic AMP/metabolism , Female , GC Rich Sequence/physiology , Humans , Molecular Sequence Data , Molecular Weight , Nuclear Proteins/metabolism , Phosphorylation , Pregnancy Proteins/chemistry , Pregnancy Proteins/metabolism , Pregnancy-Specific beta 1-Glycoproteins/metabolism
16.
Circ Res ; 91(10): 899-906, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12433834

ABSTRACT

Angiotensin-converting enzyme (ACE) is an enzyme that plays a major role in vasoactive peptide metabolism, and it has been implicated in various cardiovascular diseases. Phorbol 12-myristate 13-acetate (PMA), a protein kinase C activator, has been shown to increase ACE mRNA at the transcriptional level in human umbilical vein endothelial cells. We have investigated the transcriptional mechanism involved in protein kinase C induction of the ACE gene. Deletion and transfection analyses have revealed that two regions are required for PMA-inducible gene expression. The first is a G+C-rich region located in the proximal ACE promoter bearing overlapping consensus recognition sequences for stimulatory protein-1 (Sp1) and early growth response gene 1 (Egr-1). Electrophoretic mobility shift assay and supershift experiments have shown that Egr-1 is present in the specific nucleoprotein complex induced by PMA in human umbilical vein endothelial cells. The second region is located in the distal ACE promoter. DNase I footprinting analysis restricted this region to a 21-bp element containing a cAMP-responsive element/12-O-tetradecanoylphorbol 13-acetate-responsive element sequence. Electrophoretic mobility shift assays and supershift analyses have revealed that activating protein 1 (AP-1) is the transcription factor binding the cAMP-responsive element/12-O-tetradecanoylphorbol 13-acetate-responsive element located in the ACE promoter after PMA stimulation. Mutations of either Egr-1 or AP-1 binding sites partially abrogate ACE expression induced by PMA, whereas mutation of both sites totally abrogates PMA-induced ACE expression. Treatment of cells with PD98059, a mitogen-activated protein kinase kinase-1-specific inhibitor, inhibited PMA-induced ACE expression. Our results demonstrate that the two transcription factors, Egr-1 and AP-1, are involved in the PMA-induced ACE transcriptional activation in human endothelial cells via the activation of the extracellular signal-regulated kinase 1/2 signaling pathway.


Subject(s)
DNA-Binding Proteins/metabolism , Endothelium, Vascular/metabolism , Enzyme Induction/drug effects , Immediate-Early Proteins , Peptidyl-Dipeptidase A/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Cell Nucleus/metabolism , Cells, Cultured , DNA Footprinting , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Electrophoretic Mobility Shift Assay , Endothelium, Vascular/drug effects , Enzyme Activators/pharmacology , Enzyme Induction/physiology , Enzyme Inhibitors/pharmacology , GC Rich Sequence/physiology , Gene Expression Regulation/physiology , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis, Site-Directed , Peptidyl-Dipeptidase A/biosynthesis , Promoter Regions, Genetic/physiology , Protein Binding/physiology , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Structure-Activity Relationship , Transcription Factors/genetics , Transcription, Genetic/drug effects , Transfection
17.
Biochemistry ; 41(42): 12771-84, 2002 Oct 22.
Article in English | MEDLINE | ID: mdl-12379120

ABSTRACT

In the present study we establish that specific members of the Sp1 family of transcription factors (Sp1 and Sp3) bind to all six GC-rich motifs (elements 1-6) present in the proximal promoter of the human cell cycle inhibitor p21(WAF-1/Cip1) gene. Competition analysis showed that Sp1 and Sp3 bound with high affinity to elements 1, 3, 4, and 5/6 and with lower affinity to element 2. Transfection experiments in the Sp1-deficient Drosophila SL2 cells established that Sp1 and Sp3 but not Sp2 are potent transactivators of the p21 promoter. Transactivation by Sp1 was compromised either by deletion of element 1 (-119/-114) or by using a truncated Sp1 form lacking the C-terminal regulatory domain D. Point mutagenesis of the -2325/+8 p21 promoter, targeting individual elements 1-6, showed that mutations in element 3 (-82/-77) caused a dramatic reduction (90%) in p21 promoter activity whereas mutations in other elements had a less severe effect. The mutations in element 3 abolished p21 promoter induction by upstream enhancer elements in HepG2 cells. Sp1, but not Sp3, mediated the transactivation of the p21 promoter by the TGFbeta signaling mediator Smad3 and Smad4 proteins whereas none of the individual mutations in elements 1-6 affected the transactivation of the p21 promoter by Smad proteins in HepG2 cells. Our results suggest that functional interactions between Sp1 family members bound to specific elements of the proximal promoter and factors bound to distal enhancer elements govern the hepatic activity of the human p21 promoter under basal or inducible conditions.


Subject(s)
5' Untranslated Regions/physiology , Cyclins/genetics , Enhancer Elements, Genetic/physiology , GC Rich Sequence/physiology , Growth Inhibitors/genetics , Multigene Family , Promoter Regions, Genetic , Sp1 Transcription Factor/physiology , Animals , Base Sequence , Binding Sites/genetics , COS Cells , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Drosophila/genetics , Gene Expression Regulation/physiology , Growth Inhibitors/metabolism , Humans , Molecular Sequence Data , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Rats , Sequence Deletion , Smad Proteins , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor , Trans-Activators/metabolism , Trans-Activators/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology , Transfection , Tumor Cells, Cultured
18.
Leukemia ; 16(9): 1755-62, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12200691

ABSTRACT

PLZF (promyelocytic leukemia zinc finger ) is a transcription factor disrupted in t(11;17)-associated acute promyelocytic leukemia which is highly expressed in undifferentiated myeloid cells. To address the tissue-specific regulation of the promoter, we isolated sequences 1.2-kb 5' to the transcriptional start site. Sequence analysis demonstrated that this region contains one TATA box and several putative transcription factor binding sites including four G/C-rich sites and one Evi-1-like site. A fragment of the promoter spanning 158-bp upstream of the transcription start site displayed relative specificity for PLZF-expressing myeloid cells. Functional promoter assays revealed that an Evi-1-like site at -140/-130 was essential for full promoter activity in every cell line tested while a G-rich site at -15/-7 was important for tissue specificity. Electrophoretic mobility shift assays showed that Evi-1 binds specifically to -140/-130 Evi-1-like site and overexpression of Evi-1 in K562 cells activated the PLZF promoter. UV cross-linking assays showed that the proximal, tissue specific element at -15/-7 bound a novel 28 kDa protein. These results indicate as with other myeloid genes, a relatively small segment of DNA can direct tissue-specific expression, but unlike other myeloid promoters, no critical PU.1 or C/EBP sites were found.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , DNA/metabolism , GC Rich Sequence/physiology , Leukemia, Promyelocytic, Acute/genetics , Proto-Oncogenes , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Base Sequence , Binding Sites , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Gene Deletion , Gene Expression Regulation, Neoplastic , Humans , Kruppel-Like Transcription Factors , Leukemia, Promyelocytic, Acute/metabolism , Luciferases/metabolism , MDS1 and EVI1 Complex Locus Protein , Molecular Sequence Data , Promoter Regions, Genetic , Promyelocytic Leukemia Zinc Finger Protein , Translocation, Genetic , Tumor Cells, Cultured , Zinc Fingers
20.
J Biochem ; 130(6): 885-91, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11726291

ABSTRACT

The 5'-flanking region of the mouse Hex gene was examined in order to identify transcription factors regulating its expression in hepatocytes and haematopoietic cells. We have identified two further GC boxes (GC boxes 3 and 4 at nucleotide positions -149 to -140 and -79 to -70, respectively), i.e. in addition to the two previously determined ones (GC boxes 1 and 2 at nucleotide positions -197 to -188 and -176 to -167, respectively). Luciferase reporter assays revealed that all four GC boxes are transcriptionally active in both MH(1)C(1) rat hepatoma and K562 human chronic myelogenous leukemia cells. Electrophoretic mobility shift assays with specific competitors and antibodies showed that members of the Sp family, namely Sp1 and Sp3, bind to these GC boxes. Overexpression of Sp1 and Sp3 in Drosophila SL2 cells stimulated transcription of the Hex gene through interactions with GC boxes 1 to 4, Sp1 being a more potent activator than Sp3. Thus, we conclude that Sp1 and Sp3 stimulate transcription of the Hex gene in both MH(1)C(1) and K562 cells.


Subject(s)
DNA-Binding Proteins/physiology , GC Rich Sequence/physiology , Homeodomain Proteins/genetics , Sp1 Transcription Factor/physiology , Transcription Factors/physiology , Transcription, Genetic , Animals , Carcinoma, Hepatocellular , Cell Line , Drosophila , Genes, Homeobox , Genes, Regulator , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Promoter Regions, Genetic , Rats , Sp3 Transcription Factor , Transcriptional Activation/physiology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...