Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncol Rep ; 35(6): 3371-86, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27108960

RESUMO

Mutations in the epidermal growth factor receptor (EGFR) make lung adenocarcinoma cells sensitive to EGFR tyrosine kinase inhibitors (TKIs). Long-term cancer therapy may cause the occurrence of acquired resistance to EGFR TKIs. Long non-coding RNAs (lncRNAs) play important roles in tumor formation, tumor metastasis and the development of EGFR-TKI resistance in lung cancer. To gain insight into the molecular mechanisms of EGFR-TKI resistance, we generated an EGFR-TKI-resistant HCC827-8-1 cell line and analyzed expression patterns by lncRNA microarray and compared it with its parental HCC827 cell line. A total of 1,476 lncRNA transcripts and 1,026 mRNA transcripts were dysregulated in the HCC827­8-1 cells. The expression levels of 7 chosen lncRNAs were validated by real-time quantitative PCR. As indicated by functional analysis, several groups of lncRNAs may be involved in the bio-pathways associated with EGFR-TKI resistance through their cis- and/or trans­regulation of protein-coding genes. Thus, lncRNAs may be used as novel candidate biomarkers and potential targets in EGFR-TKI therapy in the future.


Assuntos
Adenocarcinoma/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , RNA Longo não Codificante/análise , Adenocarcinoma/genética , Adenocarcinoma de Pulmão , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Fator de Transcrição E2F1/fisiologia , Humanos , Neoplasias Pulmonares/genética , MicroRNAs/análise , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real , Fatores Estimuladores Upstream/fisiologia
2.
Circ Res ; 117(2): 142-56, 2015 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-26034040

RESUMO

RATIONALE: Wnt signaling regulates key aspects of diabetic vascular disease. OBJECTIVE: We generated SM22-Cre;LRP6(fl/fl);LDLR(-/-) mice to determine contributions of Wnt coreceptor low-density lipoprotein receptor-related protein 6 (LRP6) in the vascular smooth muscle lineage of male low-density lipoprotein receptor-null mice, a background susceptible to diet (high-fat diet)-induced diabetic arteriosclerosis. METHODS AND RESULTS: As compared with LRP6(fl/fl);LDLR(-/-) controls, SM22-Cre;LRP6(fl/fl);LDLR(-/-) (LRP6-VKO) siblings exhibited increased aortic calcification on high-fat diet without changes in fasting glucose, lipids, or body composition. Pulse wave velocity (index of arterial stiffness) was also increased. Vascular calcification paralleled enhanced aortic osteochondrogenic programs and circulating osteopontin (OPN), a matricellular regulator of arteriosclerosis. Survey of ligands and Frizzled (Fzd) receptor profiles in LRP6-VKO revealed upregulation of canonical and noncanonical Wnts alongside Fzd10. Fzd10 stimulated noncanonical signaling and OPN promoter activity via an upstream stimulatory factor (USF)-activated cognate inhibited by LRP6. RNA interference revealed that USF1 but not USF2 supports OPN expression in LRP6-VKO vascular smooth muscle lineage, and immunoprecipitation confirmed increased USF1 association with OPN chromatin. ML141, an antagonist of cdc42/Rac1 noncanonical signaling, inhibited USF1 activation, osteochondrogenic programs, alkaline phosphatase, and vascular smooth muscle lineage calcification. Mass spectrometry identified LRP6 binding to protein arginine methyltransferase (PRMT)-1, and nuclear asymmetrical dimethylarginine modification was increased with LRP6-VKO. RNA interference demonstrated that PRMT1 inhibits OPN and TNAP, whereas PRMT4 supports expression. USF1 complexes containing the histone H3 asymmetrically dimethylated on Arg-17 signature of PRMT4 are increased with LRP6-VKO. Jmjd6, a demethylase downregulated with LRP6 deficiency, inhibits OPN and TNAP expression, USF1: histone H3 asymmetrically dimethylated on Arg-17 complex formation, and transactivation. CONCLUSIONS: LRP6 restrains vascular smooth muscle lineage noncanonical signals that promote osteochondrogenic differentiation, mediated in part via USF1- and arginine methylation-dependent relays.


Assuntos
Arteriosclerose/prevenção & controle , Calcinose/prevenção & controle , Diabetes Mellitus Experimental/complicações , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/metabolismo , Receptores de LDL/deficiência , Via de Sinalização Wnt , Animais , Arginina/análogos & derivados , Arginina/metabolismo , Arteriosclerose/etiologia , Arteriosclerose/metabolismo , Calcinose/etiologia , Calcinose/metabolismo , Diabetes Mellitus Experimental/patologia , Gorduras na Dieta/efeitos adversos , Receptores Frizzled/fisiologia , Regulação da Expressão Gênica/fisiologia , Histonas/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/deficiência , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Masculino , Camundongos , Camundongos Knockout , Miócitos de Músculo Liso/patologia , Osteopontina/biossíntese , Osteopontina/genética , Comunicação Parácrina , Mapeamento de Interação de Proteínas , Proteína-Arginina N-Metiltransferases/metabolismo , Receptores de Superfície Celular , Receptores de LDL/genética , Fatores Estimuladores Upstream/fisiologia , Rigidez Vascular/fisiologia
3.
Reproduction ; 149(2): 203-12, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25385722

RESUMO

Upstream stimulating factor 1 (USF1) is a basic helix-loop-helix transcription factor that specifically binds to E-box DNA motifs, known cis-elements of key oocyte expressed genes essential for oocyte and early embryonic development. However, the functional and regulatory role of USF1 in bovine oocyte and embryo development is not understood. In this study, we demonstrated that USF1 mRNA is maternal in origin and expressed in a stage specific manner during the course of oocyte maturation and preimplantation embryonic development. Immunocytochemical analysis showed detectable USF1 protein during oocyte maturation and early embryonic development with increased abundance at 8-16-cell stage of embryo development, suggesting a potential role in embryonic genome activation. Knockdown of USF1 in germinal vesicle stage oocytes did not affect meiotic maturation or cumulus expansion, but caused significant changes in mRNA abundance for genes associated with oocyte developmental competence. Furthermore, siRNA-mediated depletion of USF1 in presumptive zygote stage embryos demonstrated that USF1 is required for early embryonic development to the blastocyst stage. A similar (USF2) yet unique (TWIST2) expression pattern during oocyte and early embryonic development for related E-box binding transcription factors known to cooperatively bind USF1 implies a potential link to USF1 action. This study demonstrates that USF1 is a maternally derived transcription factor required for bovine early embryonic development, which also functions in regulation of JY1, GDF9, and FST genes associated with oocyte competence.


Assuntos
Bovinos/embriologia , Desenvolvimento Embrionário/fisiologia , Oócitos/crescimento & desenvolvimento , Fatores Estimuladores Upstream/fisiologia , Animais , Técnicas de Cultura Embrionária/veterinária , Embrião de Mamíferos/química , Embrião de Mamíferos/fisiologia , Feminino , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica/veterinária , Técnicas de Maturação in Vitro de Oócitos/veterinária , Oócitos/química , Oogênese/fisiologia , RNA Mensageiro/análise , RNA Interferente Pequeno/farmacologia , Proteína 1 Relacionada a Twist/análise , Proteína 1 Relacionada a Twist/genética , Fatores Estimuladores Upstream/análise , Fatores Estimuladores Upstream/genética
4.
PLoS One ; 9(9): e107914, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25238393

RESUMO

The upstream stimulatory factor 2 (USF2) is a regulator of important cellular processes and is supposed to have also a role during tumor development. However, the knowledge about the mechanisms that control the function of USF2 is limited. The data of the current study show that USF2 function is regulated by phosphorylation and identified GSK3ß as an USF2-phosphorylating kinase. The phosphorylation sites within USF2 could be mapped to serine 155 and threonine 230. In silico analyses of the 3-dimensional structure revealed that phosphorylation of USF2 by GSK3ß converts it to a more open conformation which may influence transactivity, DNA binding and target gene expression. Indeed, experiments with GSK-3ß-deficient cells revealed that USF2 transactivity, DNA binding and target gene expression were reduced upon lack of GSK3ß. Further, experiments with USF2 variants mimicking GSK3ß phosphorylated USF2 in GSK3ß-deficient cells showed that phosphorylation of USF2 by GSK3ß did not affect cell proliferation but increased cell migration. Together, this study reports a new mechanism by which USF2 may contribute to cancerogenesis.


Assuntos
Quinase 3 da Glicogênio Sintase/fisiologia , Fatores Estimuladores Upstream/fisiologia , Sítios de Ligação , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação da Expressão Gênica , Glicogênio Sintase Quinase 3 beta , Meia-Vida , Células HeLa , Células Hep G2 , Humanos , Fosforilação , Ativação Transcricional , Fatores Estimuladores Upstream/química , Fatores Estimuladores Upstream/metabolismo
5.
PLoS One ; 8(8): e72358, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23991099

RESUMO

The HIF1- and HIF2-mediated transcriptional responses play critical roles in solid tumor progression. Despite significant similarities, including their binding to promoters of both HIF1 and HIF2 target genes, HIF1 and HIF2 proteins activate unique subsets of target genes under hypoxia. The mechanism for HIF target gene specificity has remained unclear. Using siRNA or inhibitor, we previously reported that STAT3 or USF2 is specifically required for activation of endogenous HIF1 or HIF2 target genes. In this study, using reporter gene assays and chromatin immuno-precipitation, we find that STAT3 or USF2 exhibits specific binding to the promoters of HIF1 or HIF2 target genes respectively even when over-expressed. Functionally, HIF1α interacts with STAT3 to activate HIF1 target gene promoters in a HIF1α HLH/PAS and N-TAD dependent manner while HIF2α interacts with USF2 to activate HIF2 target gene promoters in a HIF2α N-TAD dependent manner. Physically, HIF1α HLH and PAS domains are required for its interaction with STAT3 while both N- and C-TADs of HIF2α are involved in physical interaction with USF2. Importantly, addition of functional USF2 binding sites into a HIF1 target gene promoter increases the basal activity of the promoter as well as its response to HIF2+USF2 activation while replacing HIF binding site with HBS from a HIF2 target gene does not change the specificity of the reporter gene. Importantly, RNA Pol II on HIF1 or HIF2 target genes is primarily associated with HIF1α or HIF2α in a STAT3 or USF2 dependent manner. Thus, we demonstrate here for the first time that HIF target gene specificity is achieved by HIF transcription partners that are required for HIF target gene activation, exhibit specific binding to the promoters of HIF1 or HIF2 target genes and selectively interact with HIF1α or HIF2α protein.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Hipóxia/genética , Fator de Transcrição STAT3/fisiologia , Fatores Estimuladores Upstream/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Sítios de Ligação , Linhagem Celular , Genes Reporter , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Regiões Promotoras Genéticas , RNA Polimerase II/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
J Immunol ; 189(6): 3034-42, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22891280

RESUMO

Retinoic acid-related orphan receptor γT (RORγT) is the orphan nuclear receptor that regulates the development of Th17 cells and the expression of IL-17. The differentiation of Th17 cells is associated with the upregulation of RORγT mRNA, and the mechanisms regulating that process in human cells are not well understood. We investigated the transcriptional regulation of RORγT in a human lymphocytic cell line and Th17 differentiated from naive CD4+ cells from human peripheral blood. A series of experiments, including 5' deletion and in situ mutagenesis analysis of the human RORγT promoter, chromatin immunoprecipitation, and overexpression of selected transcription factors, revealed that the transcription factors upstream stimulatory factor 1 (USF-1) and USF-2 are indispensable for the transcription of RORγT in human lymphocytes. There was also upregulation of USF-1 and USF-2 during the differentiation of Th17 cells from naive CD4+ cells. In this article, we report the first analysis, to our knowledge, of the human RORγT promoter and demonstrate the role of the USF-1 and USF-2 transcription factors in regulating the expression of RORγT in human lymphocytes. Thus, USFs are important for the molecular mechanisms of Th17 differentiation, and possible changes in the expression of USFs might be of interest for inflammatory conditions with a Th17 component. Furthermore, these observations suggest a possible link between metabolic disorders in which the role of glucose-induced USF expression has already been established and autoimmune diseases in which the upregulation of RORγT is frequently detected.


Assuntos
Ativação Linfocitária/imunologia , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Fatores Estimuladores Upstream/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células HeLa , Células Hep G2 , Humanos , Células Jurkat , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/biossíntese , Regiões Promotoras Genéticas/imunologia , RNA Mensageiro/biossíntese , Transcrição Gênica/imunologia
7.
Hypertension ; 60(3): 697-704, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22777938

RESUMO

Estrogen receptor-α (ERα) plays a key role in the adaptation of increased uterine blood flow in pregnancy. Chronic hypoxia is a common stress to maternal cardiovascular homeostasis and causes increased risk of preeclampsia. Studies in pregnant sheep demonstrated that hypoxia during gestation downregulated ERα gene expression in uterine arteries. The present study tested the hypothesis that hypoxia causes epigenetic repression of the ERα gene in uterine arteries via heightened promoter methylation. Ovine ERα promoter of 2035 bp spanning from -2000 to +35 of the transcription start site was cloned. No estrogen or hypoxia-inducible factor response elements were found at the promoter. Two transcription factor binding sites, USF(-15) and Sp1(-520), containing CpG dinucleotides were identified, which had significant effects on the promoter activity. The USF element binds transcription factors USF1 and USF2, and the Sp1 element binds Sp1, as well as ERα through Sp1. Deletion of the Sp1 site abrogated 17ß-estradiol-induced increase in the promoter activity. In normoxic control sheep, CpG methylation at the Sp1 but not the USF site was significantly decreased in uterine arteries of pregnant as compared with nonpregnant animals. In pregnant sheep exposed to long-term high-altitude hypoxia, CpG methylation at both Sp1 and USF sites in uterine arteries was significantly increased. Methylation inhibited transcription factor binding and the promoter activity. The results provide evidence of hypoxia causing heightened promoter methylation and resultant ERα gene repression in uterine arteries and suggest new insights of molecular mechanisms linking gestational hypoxia to aberrant uteroplacental circulation and increased risk of preeclampsia.


Assuntos
Metilação de DNA/fisiologia , Epigênese Genética/fisiologia , Receptor alfa de Estrogênio/genética , Hipóxia/fisiopatologia , Prenhez/fisiologia , Regiões Promotoras Genéticas/fisiologia , Artéria Uterina/metabolismo , Animais , Ilhas de CpG/fisiologia , Regulação para Baixo/genética , Regulação para Baixo/fisiologia , Eclampsia/epidemiologia , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/metabolismo , Feminino , Modelos Animais , Gravidez , Fatores de Risco , Ovinos , Fatores Estimuladores Upstream/fisiologia
8.
J Immunol ; 188(5): 2266-75, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22287717

RESUMO

Activation of germline promoters is central to V(D)J recombinational accessibility, driving chromatin remodeling, nucleosome repositioning, and transcriptional read-through of associated DNA. We have previously shown that of the two TCRß locus (Tcrb) D segments, Dß1 is flanked by an upstream promoter that directs its transcription and recombinational accessibility. In contrast, transcription within the DJß2 segment cluster is initially restricted to the J segments and only redirected upstream of Dß2 after D-to-J joining. The repression of upstream promoter activity prior to Tcrb assembly correlates with evidence that suggests DJß2 recombination is less efficient than that of DJß1. Because inefficient DJß2 assembly offers the potential for V-to-DJß2 recombination to rescue frameshifted V-to-DJß1 joints, we wished to determine how Dß2 promoter activity is modulated upon Tcrb recombination. In this study, we show that repression of the otherwise transcriptionally primed 5'Dß2 promoter requires binding of upstream stimulatory factor (USF)-1 to a noncanonical E-box within the Dß2 12-recombination signal sequence spacer prior to Tcrb recombination. USF binding is lost from both rearranged and germline Dß2 sites in DNA-dependent protein kinase, catalytic subunit-competent thymocytes. Finally, genotoxic dsDNA breaks lead to rapid loss of USF binding and gain of transcriptionally primed 5'Dß2 promoter activity in a DNA-dependent protein kinase, catalytic subunit-dependent manner. Together, these data suggest a mechanism by which V(D)J recombination may feed back to regulate local Dß2 recombinational accessibility during thymocyte development.


Assuntos
Diferenciação Celular/genética , Diferenciação Celular/imunologia , DNA Intergênico/química , Rearranjo Gênico da Cadeia delta dos Receptores de Antígenos dos Linfócitos T/imunologia , Subpopulações de Linfócitos T/imunologia , Fatores Estimuladores Upstream/antagonistas & inibidores , Fatores Estimuladores Upstream/fisiologia , Regiões 5' não Traduzidas/genética , Regiões 5' não Traduzidas/imunologia , Regiões 5' não Traduzidas/efeitos da radiação , Animais , Sequência de Bases , Diferenciação Celular/efeitos da radiação , Linhagem Celular , Cobalto , DNA Intergênico/efeitos da radiação , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/efeitos da radiação , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Transdução de Sinais/efeitos da radiação , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/efeitos da radiação , Fatores Estimuladores Upstream/genética
9.
Biol Reprod ; 86(3): 77, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22116805

RESUMO

The aryl hydrocarbon receptor (AHR) mediates the toxicity of a variety of environmental chemicals. Apart from this, an understanding is emerging that the AHR has a fundamental role in female reproduction. Evidence suggests that AHR participates in regulation of follicle-stimulating hormone receptor (Fshr) transcript level in mouse ovary by binding to the promoter of this gene in vivo. The purpose of this study was to demonstrate the molecular interplay of the Fshr promoter involved in the transactivation by AHR in mouse granulosa cells. We found that AHR activates the Fshr promoter through the region from -209 to -99 bp. In this region, the importance of the E-box motif was revealed by site-directed mutagenesis followed by promoter analysis. By focusing on the DNA/protein interactions, we defined the fact that the intact E-box but not upstream transcription factor 1 (USF1), which is known to bind this motif, is necessary for AHR binding to mouse Fshr promoter. Furthermore, by constructing AHR mutants defective in DNA interaction, we confirmed the importance of DNA binding for AHR's ability to bind to and activate Fshr promoter. Collectively, the present study demonstrates that AHR modulates Fshr transactivation by its direct association through an E-box and not by recruitment via interaction with USFs. These observations suggest that although AHR and USF may respond to different signals, they compete on binding to the same E-box. Our data, together with that from one prior study suggesting involvement of E-box motif in AHR-mediated transcription, provide novel understanding of the way in which AHR may regulate its target genes through E-box sites.


Assuntos
Elementos E-Box/fisiologia , Regiões Promotoras Genéticas/fisiologia , Receptores de Hidrocarboneto Arílico/fisiologia , Receptores do FSH/fisiologia , Ativação Transcricional/fisiologia , Animais , Sequência de Bases , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Dados de Sequência Molecular , Mutação/genética , Ligação Proteica/fisiologia , Receptores de Hidrocarboneto Arílico/genética , Fatores Estimuladores Upstream/fisiologia
10.
J Cell Biochem ; 111(3): 720-6, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20626032

RESUMO

The polyphenol quercetin (Quer) represses expression of the cardiovascular disease risk factor plasminogen activator inhibitor-1 (PAI-1) in cultured endothelial cells (ECs). Transfection of PAI-1 promoter-luciferase reporter deletion constructs identified a 251-bp fragment (nucleotides -800 to -549) responsive to Quer. Two E-box motifs (CACGTG), at map positions -691 (E-box1) and -575 (E-box2), are platforms for occupancy by several members of the c-MYC family of basic helix-loop-helix leucine zipper (bHLH-LZ) proteins. Promoter truncation and electrophoretic mobility shift/supershift analyses identified upstream stimulatory factor (USF)-1 and USF-2 as E-box1/E-box2 binding factors. ECs co-transfected with a 251 bp PAI-1 promoter fragment containing the two E-box motifs (p251/luc) and a USF-2 expression vector (pUSF-2/pcDNA) exhibited reduced luciferase activity versus p251/luc alone. Overexpression of USF-2 decreased, while transfection of a dominant-negative USF construct increased, EC growth consistent with the known anti-proliferative properties of USF proteins. Quer-induced decreases in PAI-1 expression and reduced cell proliferation may contribute, at least in part, to the cardioprotective benefit associated with daily intake of polyphenols.


Assuntos
Células Endoteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Inibidor 1 de Ativador de Plasminogênio/genética , Quercetina/farmacologia , Fatores Estimuladores Upstream/fisiologia , Cardiotônicos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Elementos E-Box , Células Endoteliais/citologia , Humanos , Zíper de Leucina/genética , Fragmentos de Peptídeos , Inibidor 1 de Ativador de Plasminogênio/biossíntese
11.
Endocrinology ; 150(8): 3783-91, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19423764

RESUMO

FSH acts through the FSH receptor (FSHR) to modulate cell processes that are required to support developing spermatozoa. Within the testis, only Sertoli cells possess receptors for FSH and are the major targets for this regulator of spermatogenesis. FSH stimulation of Sertoli cells for 24-48 h is known to induce Fshr mRNA expression through an E-box motif (CACGTG) located 25 bp upstream of the transcription start site. In contrast, FSH stimulation for 8 h inhibits Fshr transcription. DNA-protein binding studies performed using nuclear extracts from Sertoli cells show that protein binding to the Fshr promoter E-box was reduced 68% after 6 h of FSH stimulation but increased 191% over basal levels after 48 h of stimulation. The proteins binding to the Fshr E-box were identified as upstream stimulatory factor (USF)-1 and -2. FSH stimulation transiently decreased USF1 levels and increased the expression of the inhibitor of DNA binding/differentiation (ID)-2 repressor protein with the same kinetics as the decreased USF/E-box interactions. Overexpression of ID2 resulted in a dose-dependent decrease in USF-driven Fshr promoter activity in the MSC-1 Sertoli cell line, and ID2 inhibited USF binding to the Fshr E-box. Together, these studies suggest that stimulation of Sertoli cells with FSH transiently decreases expression of the USF1 activator and induces accumulation of the ID2 repressor, to block USF binding to the Fshr promoter and delay activation of Fshr transcription. This FSH-regulated mechanism may explain the cyclical changes in Fshr expression that occurs in Sertoli cells in vivo.


Assuntos
Hormônio Foliculoestimulante/farmacologia , Hormônios/farmacologia , Proteína 2 Inibidora de Diferenciação/genética , Receptores do FSH/genética , Fatores Estimuladores Upstream/metabolismo , Animais , Linhagem Celular , Células Cultivadas , AMP Cíclico/farmacologia , Ensaio de Desvio de Mobilidade Eletroforética , Imunofluorescência , Expressão Gênica/efeitos dos fármacos , Proteína 2 Inibidora de Diferenciação/fisiologia , Masculino , Camundongos , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Ratos , Células de Sertoli/metabolismo , Fatores Estimuladores Upstream/fisiologia
12.
Biochim Biophys Acta ; 1791(4): 229-37, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19416648

RESUMO

We studied the transcriptional regulation of the HL gene by USF1 and USF2 in HepG2 cells. The transcriptional activity of the HL(-685/+13) promoter construct was increased up to 25-fold by co-transfection with USF1 and USF2. Silencing of USF1 by RNA interference reduced promoter activity by 30-40%. Chromatin immunoprecipitation assays showed binding of endogenous USF1 and USF2 to the proximal HL promoter region. In gel shift assays, USF1 and USF2 bound to E-boxes at -307/-312 and -510/-516, and to the TATA-Inr region. Although the -514C-->T substitution abolished in vitro USF binding to the -510/-516 E-box, the increase in HL promoter activity by USF1 and USF2 was unaffected. Deletion and mutation analysis of the HL promoter region, and insertion of multiple E-box copies in front of a heterologous promoter, revealed that upregulation by USFs was mainly mediated through the -307/-312 E-box and the TATA-Inr region. We conclude that in HepG2 cells USF1 and USF2 regulate transcriptional activity of the HL gene through their binding to the E-box at -307/-312 and the TATA-Inr region.


Assuntos
Elementos E-Box/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Lipase/genética , Regiões Promotoras Genéticas/genética , Fatores Estimuladores Upstream/fisiologia , Animais , Células COS , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Chlorocebus aethiops , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Regulação Neoplásica da Expressão Gênica , Humanos , Lipase/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
13.
J Biol Chem ; 284(28): 18851-62, 2009 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-19389701

RESUMO

How transcription factors interpret the output from signal transduction pathways to drive distinct programs of gene expression is a key issue that underpins development and disease. The ubiquitously expressed basic-helix-loop-helix leucine zipper upstream stimulating factor-1 binds E-box regulatory elements (CANNTG) to regulate a wide number of gene networks. In particular, USF-1 is a key component of the tanning process. Following UV irradiation, USF-1 is phosphorylated by the p38 stress-activated kinase on threonine 153 and directly up-regulates expression of the POMC, MC1R, TYR, TYRP-1 and DCT genes. However, how phosphorylation on Thr-153 might affect the activity of USF-1 is unclear. Here we show that, in response to DNA damage, oxidative stress and cellular infection USF-1 is acetylated in a phospho-Thr-153-dependent fashion. Phospho-acetylated USF-1 is nuclear and interacts with DNA but displays altered gene regulatory properties. Phospho-acetylated USF-1 is thus proposed to be associated with loss of transcriptional activation properties toward several target genes implicated in pigmentation process and cell cycle regulation. The identification of this critical stress-dependent USF-1 modification gives new insights into understanding USF-1 gene expression modulation associated with cancer development.


Assuntos
Regulação da Expressão Gênica , Fatores Estimuladores Upstream/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Acetilação , Sequência de Aminoácidos , Animais , Dano ao DNA , Humanos , Melanoma Experimental , Camundongos , Dados de Sequência Molecular , Estresse Oxidativo , Fosforilação , Processamento de Proteína Pós-Traducional , Treonina/química , Fatores Estimuladores Upstream/metabolismo
14.
Expert Rev Mol Med ; 11: e13, 2009 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-19397838

RESUMO

Approximately a third of patients with diabetes develop diabetic kidney disease, and diabetes is the leading cause of end-stage renal disease in most developed countries. Hyperglycaemia is known to activate genes that ultimately lead to extracellular matrix accumulation, the hallmark of diabetic nephropathy. Several transcription factors have been implicated in glucose-mediated expression of genes involved in diabetic nephropathy. This review focuses on the transcription factors upstream stimulatory factors 1 and 2 (USF1 and 2), activator protein 1 (AP-1), nuclear factor (NF)-kappaB, cAMP-response-element-binding protein (CREB), nuclear factor of activated T cells (NFAT), and stimulating protein 1 (Sp1). In response to high glucose, several of these transcription factors regulate the gene encoding the profibrotic cytokine transforming growth factor beta, as well as genes for a range of other proteins implicated in inflammation and extracellular matrix turnover, including thrombospondin 1, the chemokine CCL2, osteopontin, fibronectin, decorin, plasminogen activator inhibitor 1 and aldose reductase. Identifying the molecular mechanisms by which diabetic nephropathy occurs has important clinical implications as therapies can then be tailored to target those at risk. Strategies to specifically target transcription factor activation and function may be employed to halt the progression of diabetic nephropathy.


Assuntos
Nefropatias Diabéticas/fisiopatologia , Regulação da Expressão Gênica , Fatores de Transcrição/fisiologia , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/genética , Matriz Extracelular/metabolismo , Humanos , Hiperglicemia/complicações , Modelos Biológicos , Fator de Transcrição AP-1/fisiologia , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Fatores Estimuladores Upstream/fisiologia
15.
Biochim Biophys Acta ; 1791(1): 39-52, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18983939

RESUMO

Glycerol-3-phosphate acyltransferase 1 (GPAT1) is a rate limiting enzyme in de novo glycerophospholipid synthesis. The murine GPAT1 promoter sequence (the "classical" sequence) was reported previously. However, the organization of this DNA sequence does not fully match the mouse genome sequences on NCBI/GenBank. Here we have identified net cis-acting promoter sequences for the mouse GPAT1 gene: promoter 1a which includes part of the classical sequence and the downstream promoter 1b. Promoter 1a facilitates transcription of two alternative GPAT1 transcript variants, GPAT1-V1 and V2, while promoter 1b produces a third transcript variant, GPAT1-V3. Upstream stimulating factor-1 (USF-1) controlled both promoters whereas sterol regulatory element-binding protein-1 (SREBP-1) exclusively regulated promoter 1a activity in vitro. Feeding increased GPAT1-V1 and V2, but not V3 mRNA levels in mouse liver. The obese condition of db/db mice did not alter the hepatic expression levels of any of the three GPAT1 variants. Feeding enhanced hepatic mRNA levels, intranuclear protein levels and promoter 1a-binding levels of SREBP-1, but not of USF-1. Thus, promoter 1a was exclusively activated by routine feeding in vivo. Our results indicate differential roles of the two promoters in the regulation of hepatic GPAT1 gene expression in mice.


Assuntos
Glicerol-3-Fosfato O-Aciltransferase/genética , Regiões Promotoras Genéticas/genética , Animais , Sequência de Bases , Linhagem Celular Tumoral , Regulação Enzimológica da Expressão Gênica , Hepatócitos/metabolismo , Humanos , Masculino , Camundongos , Camundongos Obesos , Interferência de RNA , Proteína de Ligação a Elemento Regulador de Esterol 1/fisiologia , Fatores Estimuladores Upstream/fisiologia
16.
Am J Physiol Gastrointest Liver Physiol ; 295(6): G1211-6, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18845576

RESUMO

Human organic cation transporter 1 (OCT1/SLC22A1) plays important roles in the hepatic uptake of cationic drugs. The functional characteristics of this transporter have been well evaluated, but molecular information regarding transcriptional regulation is limited. In the present study, therefore, we examined the gene regulation of OCT1 gene focusing on basal core expression. An approximately 2.5-kb fragment of the OCT1 promoter region was isolated, and promoter activity was measured by luciferase assay in the human liver cell lines Huh7 and HepG2. Deletion analysis suggested that the region spanning -141/-69 was essential for the basal core transcriptional activity and that this region contained the sequence of a cognate E-box (CACGTG). The E-box is known to be bound by the basal transcription factors, upstream stimulating factors (USFs), and the functional involvements of USF1 and USF2 were confirmed by a transactivation effect, a mutational analysis of the E-box, and an electrophoretic mobility shift assay. The transactivation effect of USFs on the OCT1 promoter was further stimulated by hepatocyte nuclear factor 4alpha, a liver-enriched transcription factor. There were no polymorphisms in the proximal promoter region (about 400 bp) of OCT1 gene (n = 109). These findings indicated that both USF1 and USF2 bind to an E-box sequence located in the OCT1 core promoter region and are required for the basal gene expression of this transporter.


Assuntos
Fator 1 de Transcrição de Octâmero/genética , Regiões Promotoras Genéticas/genética , Fatores Estimuladores Upstream/fisiologia , Sequência de Bases , Linhagem Celular Tumoral , Análise Mutacional de DNA , Elementos E-Box/genética , Regulação da Expressão Gênica , Fator 4 Nuclear de Hepatócito , Humanos , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único , Sítio de Iniciação de Transcrição
17.
J Pediatr Surg ; 43(11): 2016-23, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18970934

RESUMO

BACKGROUND: Hepcidin is downregulated during the progression of biliary atresia (BA), but the mechanism is still unknown. METHODS: We analyzed single nucleotide polymorphism of rs7251432 and 916145 within hepcidin and its upstream, USF2 gene, respectively, in 52 patients of BA and 96 healthy controls. Liver tissues were obtained from 10 patients with early and late stage of BA, 10 patients with choledochal cyst, and 4 normal controls to study upstream stimulatory factor 2 (USF2) messenger RNA (mRNA) and protein expressions. Chromatin immunoprecipitation assay and USF2-specific short interference RNA (siRNA) were used in human HepG2 cells to show that USF2 can regulate hepcidin expression. RESULTS: C and CC allele frequencies of rs916145 of USF2 were significantly higher in patients with BA than in healthy controls. There was also significantly higher USF2 protein nuclear translocation in the early stage of BA than in the late stage, which was compatible with higher hepcidin mRNA expression in the early stage of BA. Chromatin immunoprecipitation assay demonstrated physiologic bindings of USF2 to the hepcidin promoter in HepG2 cells. USF2 siRNA also significantly knocked down hepcidin mRNA expression. CONCLUSION: The study demonstrates that C allele of rs916145 in USF2 gene has more frequency for developing BA, and decreased USF2 protein nuclear translocation might partly play a role in the decreased hepcidin expression in the cholestatic liver injury of the late stage of BA.


Assuntos
Peptídeos Catiônicos Antimicrobianos/genética , Atresia Biliar/genética , Regulação da Expressão Gênica , Fatores Estimuladores Upstream/fisiologia , Transporte Ativo do Núcleo Celular , Alelos , Peptídeos Catiônicos Antimicrobianos/biossíntese , Atresia Biliar/metabolismo , Linhagem Celular Tumoral , Pré-Escolar , Cisto do Colédoco/metabolismo , Imunoprecipitação da Cromatina , Progressão da Doença , Feminino , Frequência do Gene , Técnicas de Silenciamento de Genes , Hepatócitos/metabolismo , Hepcidinas , Humanos , Lactente , Masculino , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Fatores Estimuladores Upstream/biossíntese , Fatores Estimuladores Upstream/genética
18.
Brain Res ; 1236: 8-15, 2008 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-18755167

RESUMO

The neuron-specific K-Cl cotransporter (KCC2) maintains a low intracellular Cl(-) concentration in neurons and is necessary for fast hyperpolarizing responses to GABA and glycine. The mammalian KCC2 gene (alias Slc12a5) generates two neuron-specific isoforms by using alternative promoters and first exons. Expression of the major isoform, KCC2b, is strongly upregulated during neuronal maturation, and is modulated by neuronal activity, trauma, and neurotrophic factors. In the present study, we have focused on the regulatory influence of the upstream stimulating factors USF1 and USF2 via an E-box control element in the KCC2b promoter (E-boxKCC2b). Electrophoretic mobility shift assay in cell lines and chromatin immunoprecipitation in neurons demonstrated binding of endogenous USF1 and USF2 to the E-box(KCC2b) element. Mutation of the E-boxKCC2b site resulted in reduced KCC2b promoter activity in cell lines and cortical neurons. Overexpression of a dominant-negative form of USF confirmed the involvement of endogenous USF proteins in the regulation of the KCC2b gene. The results suggest that binding of USF proteins to the E-boxKCC2b may contribute to the upregulation of KCC2b gene expression in developing brain.


Assuntos
Elementos E-Box/fisiologia , Neurônios/metabolismo , Simportadores/metabolismo , Regulação para Cima/fisiologia , Fatores Estimuladores Upstream/fisiologia , Animais , Células Cultivadas , Córtex Cerebral/citologia , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Embrião de Mamíferos , Luciferases/biossíntese , Luciferases/genética , Camundongos , Ratos , Simportadores/genética , Transfecção , Cotransportadores de K e Cl-
19.
Am J Physiol Renal Physiol ; 295(1): F165-70, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18508876

RESUMO

Human organic cation transporter 2 (OCT2/SLC22A2), which is specifically expressed in the kidney, plays critical roles in the renal secretion of cationic compounds. Tissue expression and membrane localization of OCT2 are closely related to the tissue distribution, pharmacological effects, and/or adverse effects of its substrate drugs. However, the molecular mechanisms underlying the kidney-specific expression of OCT2 have not been elucidated. In the present study, therefore, we examined the contribution of DNA methylation of the promoter region for the OCT2 gene to its tissue-specific expression using human tissue samples. In vivo methylation status of the proximal promoter region of OCT2 and that of OCT1, a liver-specific organic cation transporter, were investigated by bisulfite sequencing using human genomic DNA extracted from the kidney and liver. All CpG sites in the OCT2 proximal promoter were hypermethylated in the liver, while hypomethylated in the kidney. On the other hand, the promoter region of OCT1 was hypermethylated in both the kidney and liver. The level of methylation of the OCT2 promoter was especially low at the CpG site in the E-box, the binding site of the basal transcription factor upstream stimulating factor (USF) 1. In vitro methylation of the OCT2 proximal promoter dramatically reduced the transcriptional activity, and an electrophoretic mobility shift assay showed that methylation at the E-box inhibited the binding of USF1. These results indicate that kidney-specific expression of human OCT2 is regulated by methylation of the proximal promoter region, interfering with the transactivation by USF1.


Assuntos
Metilação de DNA , Regulação da Expressão Gênica/fisiologia , Rim/metabolismo , Proteínas de Transporte de Cátions Orgânicos/biossíntese , Feminino , Humanos , Fígado/metabolismo , Masculino , Fator 1 de Transcrição de Octâmero/metabolismo , Transportador 2 de Cátion Orgânico , Regiões Promotoras Genéticas/efeitos dos fármacos , Fatores Estimuladores Upstream/fisiologia
20.
Mol Endocrinol ; 22(4): 904-14, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18165439

RESUMO

Local estrogen biosynthesis is a major factor in the pathogenesis of endometriosis. Aberrant expression of steroidogenic acute regulatory protein (StAR) and aromatase in endometriotic tissue leads to an up-regulation of estrogen production. The transcription factor steroidogenic factor-1 (SF-1) activates the promoters of both StAR and aromatase in endometriotic tissue. We investigated differences in SF-1 expression in endometriotic tissue and normally located endometrium to elucidate the mechanism underlying increased StAR and aromatase activities in endometriosis. Serial deletion and site-directed mutants of the SF-1 promoter showed that an E-box sequence was critical for its activity in endometriotic stromal cells. EMSAs showed that the upstream stimulatory factor (USF) 1 and 2 in nuclear extracts from endometrial and endometriotic stromal cells bound to the E-box. Chromatin-immunoprecipitation-PCR assay, however, demonstrated in intact cells that binding activity of USF2 to the SF-1 promoter was strikingly higher than that of USF1 in endometriotic stromal cells and that USF1 or USF2 binding activity was hardly detectable in endometrial stromal cells. Moreover, knockdown of USF2 but not USF1 resulted in robust and consistent down-regulation of SF-1 and its target genes StAR and aromatase in endometriotic stromal cells. USF2 but not USF1 mRNA and protein levels were significantly higher in endometriotic vs. endometrial stromal cells. In vivo, USF2 mRNA and immunoreactive USF2 levels in endometriotic tissues were strikingly higher than those in endometrium. Taken together, the elevated levels of USF2 in endometriosis account for, in part, the aberrant expression of SF-1 and its target gene StAR and aromatase.


Assuntos
Endometriose/genética , Expressão Gênica , Fator Esteroidogênico 1/genética , Fatores Estimuladores Upstream/fisiologia , Aromatase/genética , Aromatase/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina , Endometriose/metabolismo , Endometriose/patologia , Endométrio/metabolismo , Endométrio/patologia , Feminino , Humanos , Immunoblotting , Imuno-Histoquímica , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator Esteroidogênico 1/metabolismo , Células Estromais/metabolismo , Transcrição Gênica/genética , Transfecção , Fatores Estimuladores Upstream/genética , Fatores Estimuladores Upstream/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...