Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Adv Exp Med Biol ; 1395: 243-248, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36527644

RESUMO

Extracellular acidosis is a characteristic of solid tumours, resulting from hypoxia-induced glycolytic metabolism as well as from the "Warburg effect" (aerobic glycolysis). The acidic environment has shown to affect functional tumour properties (proliferation, migration, invasion) and thus the aim of the study was to identify signalling mechanisms, mediating these pH-dependent effects. Therefore, the serum response factor (Srf) and the activation of the serum response element (SRE) by acidosis were analysed in AT-1 prostate carcinoma cells. Furthermore, the expression of downstream targets of this cascade, namely the early growth response 1 (Egr1), which seems to be involved in tumour proliferation, and the cellular communication network factor 1 (Ccn1), which both contain SRE in their promotor region were examined in two tumour cell lines. Extracellular acidification led to an upregulation of Srf and a functional activation of the SRE. Egr1 expression was increased by acidosis in AT-1 cells whereas hypoxia had a suppressive effect. In experimental tumours, in vivo Egr1 and Ccn1 were also found to be acidosis-dependent. Also, it turned out that pH regulated expression of Egr1 was followed by comparable changes of p21, which is an important regulator of the cell cycle.This study identifies the Srf-SRE signalling cascade and downstream Egr1 and Ccn1 to be acidosis-regulated in vitro and in vivo, potentially affecting tumour progression. Especially linked expression changes of Egr1 and p21 may mediate acidosis-induced effects on cell proliferation.


Assuntos
Acidose , Hipóxia , Neoplasias da Próstata , Animais , Humanos , Masculino , Acidose/genética , Acidose/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/farmacologia , Hipóxia/genética , Hipóxia/metabolismo , Neoplasias Experimentais , Ativação Transcricional , Ratos , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Elemento de Resposta Sérica/genética , Elemento de Resposta Sérica/fisiologia
2.
J Biol Chem ; 291(48): 25227-25238, 2016 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-27765816

RESUMO

PLEKHG2/FLJ00018 is a Gßγ-dependent guanine nucleotide exchange factor for the small GTPases Rac and Cdc42 and has been shown to mediate the signaling pathways leading to actin cytoskeleton reorganization. Here we showed that the zinc finger domain-containing protein four-and-a-half LIM domains 1 (FHL1) acts as a novel interaction partner of PLEKHG2 by the yeast two-hybrid system. Among the isoforms of FHL1 (i.e. FHL1A, FHL1B, and FHL1C), FHL1A and FHL1B interacted with PLEKHG2. We found that there was an FHL1-binding region at amino acids 58-150 of PLEKHG2. The overexpression of FHL1A but not FHL1B enhanced the PLEKHG2-induced serum response element-dependent gene transcription. The co-expression of FHL1A and Gßγ synergistically enhanced the PLEKHG2-induced serum response element-dependent gene transcription. Increased transcription activity was decreased by FHL1A knock-out with the CRISPR/Cas9 system. Compared with PLEKHG2-expressing cells, the number and length of finger-like protrusions were increased in PLEKHG2-, Gßγ-, and FHL1A-expressing cells. Our results provide evidence that FHL1A interacts with PLEKHG2 and regulates cell morphological change through the activity of PLEKHG2.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas com Domínio LIM/metabolismo , Proteínas Musculares/metabolismo , Elemento de Resposta Sérica/fisiologia , Transcrição Gênica/fisiologia , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas com Domínio LIM/genética , Proteínas Musculares/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
3.
J Biol Chem ; 287(4): 2459-67, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22157009

RESUMO

Smooth muscle cell (SMC) differentiation is defined largely by a number of cell-restricted genes governed directly by the serum response factor (SRF)/myocardin (MYOCD) transcriptional switch. Here, we describe a new SRF/MYOCD-dependent, SMC-restricted gene known as Leiomodin 1 (Lmod1). Conventional and quantitative RT-PCRs indicate that Lmod1 mRNA expression is enriched in SMC-containing tissues of the mouse, whereas its two paralogs, Lmod2 and Lmod3, exhibit abundant expression in skeletal and cardiac muscle with very low levels in SMC-containing tissues. Western blotting and immunostaining of various adult and embryonic mouse tissues further confirm SMC-specific expression of the LMOD1 protein. Comparative genomic analysis of the human LMOD1 and LMOD2 genes with their respective mouse and rat orthologs shows high conservation between the three exons and several noncoding sequences, including the immediate 5' promoter region. Two conserved CArG boxes are present in both the LMOD1 and LMOD2 promoter regions, although LMOD1 displays much higher promoter activity and is more responsive to SRF/MYOCD stimulation. Gel shift assays demonstrate clear binding between SRF and the two CArG boxes in human LMOD1. Although the CArG boxes in LMOD1 and LMOD2 are similar, only LMOD1 displays SRF or MYOCD-dependent activation. Transgenic mouse studies reveal wild type LMOD1 promoter activity in cardiac and vascular SMC. Such activity is abolished upon mutation of both CArG boxes. Collectively, these data demonstrate that Lmod1 is a new SMC-restricted SRF/MYOCD target gene.


Assuntos
Autoantígenos/biossíntese , Diferenciação Celular/fisiologia , Proteínas do Citoesqueleto/biossíntese , Regulação da Expressão Gênica/fisiologia , Proteínas Musculares/biossíntese , Miócitos de Músculo Liso/metabolismo , Elemento de Resposta Sérica/fisiologia , Fator de Resposta Sérica/imunologia , Animais , Autoantígenos/genética , Células COS , Chlorocebus aethiops , Proteínas do Citoesqueleto/genética , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/biossíntese , Proteínas dos Microfilamentos/genética , Proteínas Musculares/genética , Miócitos de Músculo Liso/citologia , Células NIH 3T3 , Especificidade de Órgãos/fisiologia , Ratos , Fator de Resposta Sérica/genética
4.
Biol Pharm Bull ; 36(7): 1204-7, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23811570

RESUMO

FLJ00018, a heterotrimeric guanosine 5'-triphosphate (GTP)-binding protein (G protein) Gßγ subunit-activated guanine nucleotide exchange factor for Rho family small GTPases, regulates cellular responses, including cell morphological changes and gene transcriptional regulation, and targets the cellular membranes. FLJ00018 contains a Dbl homology (DH) domain in addition to a pleckstrin homology (PH) domain. Here we show that the PH domain of FLJ00018 is required for FLJ00018-induced, serum response element-dependent gene transcription. Although the PH domain of KIAA1415/P-Rex1, another Gßγ subunit-activated guanine nucleotide exchange factor for Rho family small GTPases, binds to phosphatidylinositol 3,4,5-triphosphate and phosphatidylinositol 3,4-bisphosphate, the PH domain of FLJ00018 binds to polyphosphoinositides including phosphatidylinositol 4,5-bisphosphate, and phosphatidic acid. These results suggest that FLJ00018 is targeted via its PH domain to cellular membranes.


Assuntos
Membrana Celular/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Lipídeos de Membrana/metabolismo , Fracionamento Celular , DNA Complementar/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Humanos , Luciferases/genética , Plasmídeos , Ligação Proteica , Fatores de Troca de Nucleotídeo Guanina Rho , Elemento de Resposta Sérica/fisiologia , Transcrição Gênica
5.
Endocr J ; 59(10): 867-79, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22785235

RESUMO

We examined the effects of sex steroids on prolactin promoter activity in rat somatolactotrophic GH3 cells. Both estradiol (E2) and progesterone (P4) were found to inhibit basal prolactin promoter activity, but to potentiate Thyrotropin-releasing hormone (TRH)-induced prolactin promoter activity. P4 had a greater inhibitory effect on basal prolactin promoter activity than E2, and P4 also potentiated TRH-induced prolactin promoter more potently than E2. Combined treatment with E2 and P4 further increased TRH-induced prolactin promoter activity. E2 and P4 also both reduced basal serum response element (SRE) promoter activity, and increased TRH-induced SRE promoter activity. Combination treatment with E2 and P4 reduced basal activity of SRE promoter and increased TRH-induced SRE activity more potently than E2 or P4 alone. In contrast, basal cAMP response element (CRE) promoter activity was not influenced by either E2 or P4, although TRH-induced CRE promoter was potentiated by each of these steroids, and was further increased by E2 and P4 combination treatment. Both E2 and P4 increased TRH-induced extracellular signal-regulated kinase (ERK) phosphorylation; however, intracellular cAMP levels was not influenced by E2 or P4. TRH-induced CRE promoter was inhibited by mitogen-activated protein kinase/ERK kinase (MEK) inhibitor and was increased by overexpression of MEK kinase (MEKK). This study showed that ERK and SRE transcriptional pathways, but not the cAMP/CRE pathway, may be involved in the suppression of basal prolactin promoter activity, whereas both the ERK/SRE and MAP kinase-mediated CRE pathways appear to be involved in the increased transcriptional efficiency of the prolactin promoter induced by TRH stimulation.


Assuntos
Estradiol/farmacologia , Lactotrofos/metabolismo , Progesterona/farmacologia , Prolactina/biossíntese , Animais , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Ratos , Elemento de Resposta Sérica/genética , Elemento de Resposta Sérica/fisiologia , Hormônio Liberador de Tireotropina/farmacologia
6.
J Pharmacol Exp Ther ; 332(2): 469-78, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19855098

RESUMO

G2A is a G protein-coupled receptor that can be induced by various stressors. G2A is reported to have proton-sensing activity that mediates intracellular inositol phosphate (IP) accumulation with decreasing pH. We previously showed that G2A is also activated by some oxidized free fatty acids such as 9-hydroxyoctadecadienoic acid (9-HODE). In this study, we identified a novel alternative splice variant of G2A (G2A-b) that has a partially different N terminus compared with the G2A originally reported (G2A-a). The two splice variants of G2A show similar tissue distributions, but G2A-b is expressed more abundantly. There was no difference between the two variants in 9-HODE-induced cellular responses, such as intracellular calcium mobilization and GDP/GTP exchange of Galpha protein, and in proton-sensitive IP accumulation. However, G2A-b showed a higher basal activity in terms of IP accumulation. Mutagenesis study revealed that the difference in the basal activity is attributable to the K7 residue that exists only in G2A-a. We further demonstrated that an R42A mutation largely impaired both the basal and proton-sensing activities, but did not affect the 9-HODE-induced intracellular calcium increase. Taken together, we found an additional novel G2A variant (G2A-b) that is the major transcript with functional response to ligand stimulation as well as G2A-a, and succeeded in discriminating proton-sensing and oxidized fatty acid-sensing activities of G2A.


Assuntos
Processamento Alternativo , Proteínas de Ciclo Celular/genética , Isoformas de Proteínas/genética , Receptores Acoplados a Proteínas G/genética , Sequência de Aminoácidos , Animais , Células CHO , Células COS , Cálcio/metabolismo , Proteínas de Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Chlorocebus aethiops , Cricetinae , Cricetulus , Proteína A de Ligação a Elemento de Resposta do AMP Cíclico/metabolismo , Guanosina Trifosfato/metabolismo , Células HL-60 , Humanos , Fosfatos de Inositol/metabolismo , Leucócitos , Ácidos Linoleicos Conjugados/farmacologia , Dados de Sequência Molecular , Isoformas de Proteínas/efeitos dos fármacos , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Elemento de Resposta Sérica/fisiologia , Transfecção
7.
Circ Res ; 103(1): 61-9, 2008 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-18511849

RESUMO

Lipoma preferred partner (LPP) was recently recognized as a smooth muscle marker that plays a role in smooth muscle cell migration. In this report, we focus on the transcriptional regulation of the LPP gene. In particular, we investigate whether LPP is directly regulated by serum response factor (SRF). We show that the LPP gene contains 3 evolutionarily conserved CArG boxes and that 1 of these is part of an alternative promoter in intron 2. Quantitative RT-PCR shows that this alternative promoter directs transcription specifically to smooth muscle containing tissues in vivo. By using chromatin immunoprecipitation, we demonstrate that 2 of the CArG boxes, including the promoter-associated CArG box, bind to endogenous SRF in cultured aortic smooth muscle cells. Electrophoretic mobility-shift assays show that the conserved CArG boxes bind SRF in vitro. In reporter experiments, we show that the alternative promoter has transcriptional capacity that is dependent on SRF/myocardin and that the promoter associated CArG box is required for that activity. Finally, we show by quantitative RT-PCR that the alternative promoter is strongly downregulated in SRF-deficient embryonic stem cells and in smooth muscle tissues derived from conditional SRF knockout mice. Collectively, our data demonstrate that expression of LPP in smooth muscle is mediated by an alternative promoter that is regulated by SRF/myocardin.


Assuntos
Aorta/metabolismo , Proteínas do Citoesqueleto/biossíntese , Íntrons/fisiologia , Miócitos de Músculo Liso/metabolismo , Proteínas Nucleares/metabolismo , Elemento de Resposta Sérica/fisiologia , Fator de Resposta Sérica/metabolismo , Transativadores/metabolismo , Animais , Aorta/citologia , Movimento Celular/fisiologia , Células Cultivadas , Proteínas do Citoesqueleto/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Proteínas com Domínio LIM , Masculino , Camundongos , Miócitos de Músculo Liso/citologia , Proteínas Nucleares/genética , Fator de Resposta Sérica/genética , Transativadores/genética , Transcrição Gênica/fisiologia
8.
Circ Res ; 102(12): 1502-11, 2008 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-18497331

RESUMO

Leupaxin is a LIM domain-containing adapter protein belonging to the paxillin family that has been previously reported to be preferentially expressed in hematopoietic cells. Herein, we identified leupaxin in a screen for focal adhesion kinase binding partners in aortic smooth muscle, and we show that leupaxin is enriched in human and mouse vascular smooth muscle and that leupaxin expression is dynamically regulated during development. In addition, our studies reveal that leupaxin can undergo cytoplasmic/nuclear shuttling and functions as an serum response factor cofactor in the nucleus. We found that leupaxin forms a complex with serum response factor and associates with CArG-containing regions of smooth muscle promoters and that ectopic expression of leupaxin induces smooth muscle marker gene expression in both 10T1/2 cells and rat aortic smooth muscle cells. Subsequent studies indicated that enhanced focal adhesion kinase activity (induced by fibronectin or expression of constitutively active focal adhesion kinase) attenuates the nuclear accumulation of leupaxin and limits the ability of leupaxin to enhance serum response factor-dependent gene transcription. Thus, these studies indicate that modulation of the subcellular localization of serum response factor cofactors is 1 mechanism by which extracellular matrix-dependent signals may regulate phenotypic switching of smooth muscle cells.


Assuntos
Moléculas de Adesão Celular/fisiologia , Miócitos de Músculo Liso/metabolismo , Fosfoproteínas/fisiologia , Elemento de Resposta Sérica/fisiologia , Animais , Aorta/citologia , Aorta/embriologia , Aorta/crescimento & desenvolvimento , Transporte Biológico , Moléculas de Adesão Celular/farmacologia , Diferenciação Celular , Células Cultivadas/efeitos dos fármacos , Vasos Coronários/citologia , Feminino , Quinase 1 de Adesão Focal/fisiologia , Adesões Focais/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Pessoa de Meia-Idade , Miócitos de Músculo Liso/efeitos dos fármacos , Miométrio/citologia , Especificidade de Órgãos , Fosfoproteínas/farmacologia , Mapeamento de Interação de Proteínas , Ratos , Proteínas Recombinantes de Fusão/fisiologia , Elemento de Resposta Sérica/efeitos dos fármacos , Fator de Resposta Sérica/fisiologia , Transdução de Sinais/fisiologia , Fatores de Transcrição , Transcrição Gênica
9.
J Clin Invest ; 116(1): 36-48, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16395403

RESUMO

Precise control of SMC transcription plays a major role in vascular development and pathophysiology. Serum response factor (SRF) controls SMC gene transcription via binding to CArG box DNA sequences found within genes that exhibit SMC-restricted expression. However, the mechanisms that regulate SRF association with CArG box DNA within native chromatin of these genes are unknown. Here we report that SMC-restricted binding of SRF to murine SMC gene CArG box chromatin is associated with patterns of posttranslational histone modifications within this chromatin that are specific to the SMC lineage in culture and in vivo, including methylation and acetylation to histone H3 and H4 residues. We found that the promyogenic SRF coactivator myocardin increased SRF association with methylated histones and CArG box chromatin during activation of SMC gene expression. In contrast, the myogenic repressor Kruppel-like factor 4 recruited histone H4 deacetylase activity to SMC genes and blocked SRF association with methylated histones and CArG box chromatin during repression of SMC gene expression. Finally, we observed deacetylation of histone H4 coupled with loss of SRF binding during suppression of SMC differentiation in response to vascular injury. Taken together, these findings provide novel evidence that SMC-selective epigenetic control of SRF binding to chromatin plays a key role in regulation of SMC gene expression in response to pathophysiological stimuli in vivo.


Assuntos
Cromatina/metabolismo , Regulação da Expressão Gênica , Músculo Liso Vascular/fisiologia , Elemento de Resposta Sérica/fisiologia , Fator de Resposta Sérica/metabolismo , Animais , Aorta , Sítios de Ligação , Técnicas de Cultura de Células , Heterocromatina/metabolismo , Histonas/genética , Fator 4 Semelhante a Kruppel , Músculo Liso Vascular/citologia , Ligação Proteica , Ratos , Transfecção
10.
Endocr J ; 56(3): 335-44, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19352056

RESUMO

The hypothalamic-pituitary-adrenal (HPA) axis is activated under various stressors. Corticotropin-releasing factor (CRF) plays a central role in controlling stress response, and regulating the HPA axis. CRF, produced in the hypothalamic paraventricular nucleus (PVN), stimulates adrenocorticotropic hormone (ACTH) production via CRF receptor type 1 (CRF(1) receptor) from the corticotrophs of the anterior pituitary (AP). Cyclic AMP (cAMP)-protein kinase A (PKA) pathway takes a main role in stimulating CRF gene transcription. Forskolin and pituitary adenylate cyclase-activating polypeptide (PACAP) stimulate adenylate cyclase, intracellular cAMP production, and then CRF and arginine vasopressin (AVP) gene expression in hypothalamic 4B cells. Interleukin (IL)-6, produced in the PVN, both directly and indirectly stimulates CRF and AVP gene expression. Estradiol may enhance the activation of CRF gene expression in response to stress. The HPA axis is regulated by a negative feedback mechanism, because glucocorticoids inhibit both CRF production in the hypothalamic PVN and ACTH production in the pituitary. Hypothalamic parvocellular neurons in the PVN are known to express glucocorticoid receptors, and glucocorticoids are able to regulate CRF gene transcription and expression levels directly in the PVN. Glucocorticoids-dependent repression of cAMP-stimulated CRF promoter activity is mainly localized to promoter sequences between -278 and -233 bp. Both negative glucocorticoid regulatory element (nGRE) and serum response element (SRE) are involved in the repression of the CRF gene in the hypothalamic cells.


Assuntos
Hormônio Liberador da Corticotropina/biossíntese , Regulação da Expressão Gênica , Hipotálamo/metabolismo , Animais , Arginina Vasopressina/fisiologia , Colforsina/farmacologia , Hormônio Liberador da Corticotropina/genética , AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dexametasona/farmacologia , Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Estrogênios/fisiologia , Retroalimentação , Humanos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/fisiologia , Interleucina-6/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/fisiologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Elemento de Resposta Sérica/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
11.
J Clin Invest ; 115(2): 418-27, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15690088

RESUMO

CC(A/T)6GG-dependent (CArG-dependent) and serum response factor-dependent (SRF-dependent) mechanisms are required for gene expression in smooth muscle cells (SMCs). However, an unusual feature of many SMC-selective promoter CArG elements is that they contain a conserved single G or C substitution in their central A/T-rich region, which reduces binding affinity for ubiquitously expressed SRF. We hypothesized that this CArG degeneracy contributes to cell-specific expression of smooth muscle alpha-actin in vivo, since substitution of c-fos consensus CArGs for the degenerate CArGs resulted in relaxed specificity in cultured cells. Surprisingly, our present results show that these substitutions have no effect on smooth muscle-specific transgene expression during normal development and maturation in transgenic mice. However, these substitutions significantly attenuated injury-induced downregulation of the mutant transgene under conditions where SRF expression was increased but expression of myocardin, a smooth muscle-selective SRF coactivator, was decreased. Finally, chromatin immunoprecipitation analyses, together with cell culture studies, suggested that myocardin selectively enhanced SRF binding to degenerate versus consensus CArG elements. Our results indicate that reductions in myocardin expression and the degeneracy of CArG elements within smooth muscle promoters play a key role in phenotypic switching of smooth muscle cells in vivo, as well as in mediating responses of CArG-dependent smooth muscle genes and growth regulatory genes under conditions in which these 2 classes of genes are differentially expressed.


Assuntos
Actinas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Crescimento/fisiologia , Músculo Liso/embriologia , Miócitos de Músculo Liso/metabolismo , Elemento de Resposta Sérica/fisiologia , Actinas/genética , Animais , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/genética , Crescimento/genética , Camundongos , Camundongos Transgênicos , Músculo Liso/lesões , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Especificidade de Órgãos/genética , Especificidade de Órgãos/fisiologia , Mutação Puntual/fisiologia , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Elemento de Resposta Sérica/genética , Transativadores/genética , Transativadores/metabolismo
12.
J Steroid Biochem Mol Biol ; 108(1-2): 137-48, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17942302

RESUMO

Using a luciferase reporter assay we found that human serum transactivated the ileal apical sodium-dependent bile acid transporter (ASBT) promoter three to fourfold. Confirming this effect, addition of human serum to both Caco-2 cells and fresh human ileal biopsies caused an approximate 2.0-fold increase in endogenous ASBT mRNA production. Alteration of non-esterified fatty acid (NEFA) content and cortisol content did not affect the transactivation potential of serum. Site-directed mutagenesis of response elements for corticosteroid, peroxisome proliferation-activated alpha (PPARalpha), hepatocyte nuclear factor 1alpha (HNF1alpha), and retinoic acid (RAR/RXR) did not affect transactivation potential of serum. Three putative serum response elements (SRE) were identified on the promoter, but all were determined inactive using site-directed mutagenesis and electrophoretic mobility shift assay. Promoter deletion analysis demonstrated that >80% of the response to serum was located within the last 273 bp of the 5'-UTR, an area containing one of two activate protein 1 (AP-1) response elements. Site-directed mutagenesis of this downstream AP-1 response element reduced the effect of serum on the promoter by about 50% while full deletion of the response element completely eliminated the effect of serum. These studies demonstrate that one or more constituents of human stimulate ASBT gene expression largely via the down-stream AP-1 response element.


Assuntos
Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Soro/fisiologia , Simportadores/genética , Ativação Transcricional , Animais , Células CACO-2 , Proteínas de Transporte/farmacologia , Galinhas , Corticosterona , Ácidos Graxos não Esterificados/farmacologia , Genes Reporter/efeitos dos fármacos , Humanos , Hidrocortisona/farmacologia , Íleo/metabolismo , Luciferases/genética , Mifepristona/farmacologia , Mutagênese Sítio-Dirigida , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , RNA Mensageiro/metabolismo , Receptores do Ácido Retinoico/metabolismo , Elemento de Resposta Sérica/efeitos dos fármacos , Elemento de Resposta Sérica/fisiologia , Simportadores/metabolismo , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional/efeitos dos fármacos , Transfecção , Células Tumorais Cultivadas
13.
Mol Cell Biol ; 25(11): 4792-803, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15899879

RESUMO

The mitogen-activated protein kinase (MAPK) phosphatase 1 (MKP-1) is an immediate-early gene comprised of a dual-specificity phosphatase domain and a noncatalytic NH(2) terminus. Here, we show that the NH(2) terminus of MKP-1, containing the cdc25 homology domains A (CH2A) and B (CH2B), mediates MKP-1 nuclear targeting and modulates MAPK-mediated gene expression. An LXXLL motif which is known to mediate protein-protein interactions with nuclear-targeted hormone receptors was identified proximal to the CH2A domain of MKP-1. The NH(2) terminus alone of MKP-1 containing this LXXLL motif was sufficient to direct nuclear targeting, and mutating this motif to LXXAA resulted in the exclusion of MKP-1 from the nucleus. We found that the LXXLL motif proximal to the CH2A domain was present in other nuclear-localized MKPs but was absent in MKPs that localized to the cytoplasm. These data suggest that this LXXLL motif confers nuclear targeting properties to the MKPs. The NH(2) terminus of MKP-1 was also found to inhibit the activation of the serum response element (SRE) by preventing MAPK-mediated phosphorylation of the regulatory serine 383 residue on Elk-1. Moreover, we show that MKP-1 plays a major role in the attenuation of serum-induced SRE activity, since MKP-1 null fibroblasts exhibited enhanced SRE activity in response to serum compared with wild-type fibroblasts. The NH(2) terminus of MKP-1, when reconstituted into MKP-1 null fibroblasts to levels similar to endogenous MKP-1 following serum stimulation, reduced serum-mediated SRE activity. Collectively, these data reveal novel roles for the NH(2) terminus of MKP-1 in nuclear targeting and transcriptional regulation.


Assuntos
Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Proteínas Imediatamente Precoces/química , Proteínas Imediatamente Precoces/metabolismo , Fosfoproteínas Fosfatases/química , Fosfoproteínas Fosfatases/metabolismo , Proteínas Tirosina Fosfatases/química , Proteínas Tirosina Fosfatases/metabolismo , Elemento de Resposta Sérica/fisiologia , Transcrição Gênica , Transporte Ativo do Núcleo Celular , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Células COS , Proteínas de Ciclo Celular/genética , Núcleo Celular/química , Chlorocebus aethiops , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fosfatase 1 de Especificidade Dupla , Regulação da Expressão Gênica , Proteínas Imediatamente Precoces/genética , Camundongos , Dados de Sequência Molecular , Sinais de Localização Nuclear , Fosfoproteínas Fosfatases/genética , Proteína Fosfatase 1 , Estrutura Terciária de Proteína , Proteínas Tirosina Fosfatases/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Elk-1 do Domínio ets
14.
Circ Res ; 97(4): 305-13, 2005 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-16037570

RESUMO

Atherosclerosis is considered to be an inflammatory disease. Tissue factor (TF), a prothrombotic molecule expressed by various cell types within atherosclerotic plaques, is thought to play an essential role in thrombus formation after atherosclerotic plaque rupture. Recent studies suggest that the antiinflammatory cytokine interleukin-10 (IL-10) has many antiatherosclerotic properties. Therefore, the effects of IL-10 on TF expression in response to inflammation were investigated. Mouse macrophages were stimulated with lipopolysaccharide (LPS) in the presence or absence of IL-10. Pretreatment with IL-10 resulted in a 50% decrease in TF mRNA expression and TF promoter activity. Binding of early growth response gene-1 (Egr-1) to the consensus DNA sequence, a key transcriptional activator of TF expression in response to inflammation, and the expression of Egr-1 mRNA were also inhibited by IL-10. This inhibition was independent of the induction of suppressor of cytokine signaling protein-3 by IL-10. Macrophages that had been transfected with luciferase reporter constructs containing the murine Egr-1 5'-flanking sequence exhibited reduced reporter gene activity in response to LPS stimulation with IL-10 pretreatment. Studies with deletion constructs of the Egr-1 promoter identified the proximal serum response element SRE3 as a potential regulatory site for the IL-10 mediated suppression of Egr-1 expression. Furthermore, activation of the upstream signal-transduction elements, such as mitogen-activated protein kinase kinase (MEK) 1/2, extracellular signal-regulated kinase 1/2, and Elk-1 were also inhibited by IL-10 pretreatment. Taken together, these results demonstrate a pathway for the IL-10 mediated inhibition of TF expression during inflammation and may explain the antiatherosclerotic effects of IL-10.


Assuntos
Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas Imediatamente Precoces/antagonistas & inibidores , Interleucina-10/farmacologia , Lipopolissacarídeos/farmacologia , MAP Quinase Quinase 1/fisiologia , MAP Quinase Quinase 2/fisiologia , Macrófagos/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Elemento de Resposta Sérica/fisiologia , Tromboplastina/genética , Fatores de Transcrição/antagonistas & inibidores , Animais , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteína 1 de Resposta de Crescimento Precoce , Proteínas Imediatamente Precoces/genética , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Regiões Promotoras Genéticas , RNA Mensageiro/análise , Receptores Imunológicos/fisiologia , Proteínas Repressoras/fisiologia , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina , Receptor 4 Toll-Like , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia
15.
Arterioscler Thromb Vasc Biol ; 26(5): 1029-35, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16497989

RESUMO

OBJECTIVE: Lysophosphatidic acid (LPA), one component of oxidized low-density lipoprotein, is a potent bioactive phospholipid. Early growth response gene-1 (Egr-1), an important transcription factor, regulates expression of an array of genes involved in vascular diseases. Whether and how LPA regulates the transcriptional machinery of Egr-1 gene is unknown and is addressed in this study. METHOD AND RESULTS: We found that LPA markedly induces Egr-1 mRNA and protein in aortic smooth muscle cells (SMCs). RNA stability and nuclear run-on assays reveal that LPA-induced Egr-1 gene expression is controlled at the transcriptional level. Reporter gene analyses have shown that the -141 to +20 nt region of the Egr-1 promoter contains regulatory elements. Electrophoretic mobility shift assays reveal that the DNA-binding activities of both CREB and SRF to the CRE and SRE motifs of the Egr-1 promoter are markedly elevated in response to LPA. The increased binding activity depends on the phosphorylation of CREB and SRF. Luciferase assays of a series of deleted or mutated Egr-1 promoter-reporter gene constructs, along with dominant negative CREB transfection analysis revealed that the 2 CRE sites and the 2 proximal SRE sites in the Egr-1 promoter are required for maximal LPA-induced Egr-1 gene expression. CONCLUSIONS: Our data reveal that LPA regulates Egr-1 expression via transcription factors CREB and SRF. These results establish a novel role for CREB in mediating LPA-induced gene expression. Our results imply that elevated LPA levels may, through activation of Egr-1, which regulates an array of atherogenic genes, exacerbate atheromatous lesions.


Assuntos
AMP Cíclico/farmacologia , Proteína 1 de Resposta de Crescimento Precoce/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Elementos de Resposta/fisiologia , Elemento de Resposta Sérica/fisiologia , Transcrição Gênica , Animais , Núcleo Celular/metabolismo , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Músculo Liso Vascular/citologia , Fosforilação , Regiões Promotoras Genéticas , RNA Mensageiro/análise , Ratos , Fator de Resposta Sérica/fisiologia , Transdução de Sinais
16.
Cancer Lett ; 370(1): 91-9, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26515162

RESUMO

The polyphenolic flavone chrysin has been evaluated as a natural chemopreventive agent due to its anti-cancer effects in a variety of cancer cell lines. However, the mechanism of the chemopreventive effect has been not well established, especially in human colorectal cancer cells. We evaluated the chemopreventive effect of chrysin in three different human colorectal cancer cell lines. We found that chrysin treatment consequently reduced cell viability via induction of apoptosis. We identified that the involvement of up-regulation of pro-apoptotic cytokines tumor necrosis factor (Tnf) α and ß genes and consequent activation of the TNF-mediated transcriptional pathway in chrysin-induced apoptosis. Using our generated AHR siRNA expressing colorectal cancer cells, we demonstrated that the chrysin-induced up-regulation of Tnfα and ß gene expression was dependent on the aryl hydrocarbon receptor (AHR), which is a ligand-receptor for chrysin. Subsequently, we found that the AHR siRNA expressing colorectal cancer cells were resistant to chrysin-induced apoptosis. Therefore, we concluded that AHR is required for the chrysin-induced apoptosis and the up-regulation of Tnfα and ß gene expression in human colorectal cancer cells.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Flavonoides/farmacologia , Receptores de Hidrocarboneto Arílico/fisiologia , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A2/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Linfotoxina-alfa/genética , Elemento de Resposta Sérica/fisiologia , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/fisiologia
17.
Oncogene ; 22(38): 5946-57, 2003 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-12955073

RESUMO

CrkII belongs to the adaptor protein family that plays a crucial role in signal transduction. In order to better understand the biological functions of CrkII, we focused on the regulation of gene expression by CrkII. Various transcriptional control elements were examined for their activation by CrkII-expression, and we found that CrkII selectively activates the serum response element (SRE), a transcriptional control element of immediate-early genes. This SRE activation induced by CrkII-overexpression was mediated by the serum response factor (SRF) via Rho. Indeed, we confirmed that the amount of activated Rho was increased in the CrkII-expressing cells. Moreover, we showed that when overexpressed, CrkII induces the cellular transformation of NIH 3T3 cells and that a dominant negative mutant of Rho suppresses this transformation, strongly suggesting that activation of Rho is essential for the transforming activity by CrkII. Furthermore, we also found that CrkII and Galpha12, a member of the heterotrimeric G proteins, synergistically activates Rho as well as the SRF, and that an SH3 mutant of CrkII can inhibit the Galpha12-induced activation of SRF. These results strongly suggest that CrkII is involved in the activation of Rho and SRF by Galpha12. Our study provides strong evidence that Rho activation plays a crucial role in CrkII-mediated signals to induce gene expression and cellular transformation.


Assuntos
Transformação Celular Neoplásica , Proteínas Proto-Oncogênicas/metabolismo , Elemento de Resposta Sérica/fisiologia , Fator de Resposta Sérica/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Células 3T3/metabolismo , Células 3T3/patologia , Animais , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP , Regulação da Expressão Gênica , Genes Reporter , Proteínas Heterotriméricas de Ligação ao GTP/genética , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Camundongos , Mutação , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-crk , Fator de Resposta Sérica/genética , Transdução de Sinais , Ativação Transcricional , Proteínas rho de Ligação ao GTP/genética
18.
FEBS J ; 272(12): 3105-19, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15955069

RESUMO

Serum response factor (SRF) is a MADS transcription factor that binds to the CArG box sequence of the serum response element (SRE). Through its binding to CArG sequences, SRF activates several muscle-specific genes as well as genes that respond to mitogens. The thermodynamic parameters of the interaction of core-SRF (the 124-245 fragment of serum response factor) with specific oligonucleotides from c-fos and desmin promoters, were determined by spectroscopy. The rotational correlation time of core-SRF labeled with bis-ANS showed that the protein is monomeric at low concentration (10(-7) m). The titration curves for the fluorescence anisotropy of fluorescein-labeled oligonucleotide revealed that under equilibrium conditions, the core-SRF monomers were bound sequentially to SRE at very low concentration (10(-9) m). Curve-fitting data showed also major differences between the wild-type sequence and the oligonucleotide sequences mutated within the CArG box. The fluorescence of the core-SRF tyrosines was quenched by the SRE oligonucleotide. This quenching indicated that under stoichiometric conditions, core-SRF was bound as a dimer to the wild-type oligonucleotide, and as a monomer or a tetramer to the mutant oligonucleotides. Far-UV CD spectra indicated that the flexibility of core-SRF changed profoundly upon its binding to its specific target SRE. Lastly, the rotational correlation time of fluorescein-labeled SRE revealed that formation of the specific complex was accompanied by a change in the SRE internal dynamics. These results indicated that the flexibility of the two partners is crucial for the DNA-protein interaction.


Assuntos
DNA/metabolismo , Modelos Moleculares , Elemento de Resposta Sérica/fisiologia , Fator de Resposta Sérica/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Dicroísmo Circular , DNA/química , Desmina/genética , Dimerização , Polarização de Fluorescência , Genes fos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Oligonucleotídeos/metabolismo , Regiões Promotoras Genéticas , Conformação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fator de Resposta Sérica/genética , Soluções
20.
Cell Signal ; 16(2): 201-9, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14636890

RESUMO

Heterotrimeric Galpha12/13 signals induce cellular responses such as serum response element (SRE)-mediated gene transcription via Rho GTPase. Guanine nucleotide exchange factors (GEFs) are strong candidates for linking Galpha signals to Rho. For example, p115 RhoGEF transduces Galpha13 signals to Rho and inhibits Galpha12/13 signals via the RhoGEF LH domain which links to Galpha subunits. Here, we have evaluated the signaling capacity of Lbc RhoGEF in the context of Galpha12/13 signals. In vitro GEF assays indicate that baculoviral-expressed proto-Lbc has minimal exchange activity, implying that a stimulus is required for Lbc activity in vivo. Expression of a catalytically inactive proto-Lbc mutant in HEK293T cells attenuates Galpha12- and thrombin-induced activation of an SRE transcriptional reporter, and the levels of inhibition observed is similar to that obtained with an analogous p115 RhoGEF mutant. proto-Lbc mutant expression also led to decreased levels of Galpha12-induced RhoA activation in vivo. Complex formation between Galpha12 and Lbc forms was detected. Analysis of the Lbc peptide sequence reveals a previously undetected region which may link to Galpha subunit signals. These findings support a role for Lbc in Galpha12-induced signaling pathways to Rho.


Assuntos
Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/farmacologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Elemento de Resposta Sérica/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo , Proteínas de Ancoragem à Quinase A , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Células Cultivadas , Humanos , Antígenos de Histocompatibilidade Menor , Dados de Sequência Molecular , Mutação , Estrutura Terciária de Proteína/genética , Fatores de Troca de Nucleotídeo Guanina Rho , Homologia de Sequência de Aminoácidos , Elemento de Resposta Sérica/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Trombina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA