Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Molecules ; 26(20)2021 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-34684708

RESUMO

Elk-1 is a transcription factor that binds together with a dimer of the serum response factor (SRF) to the serum-response element (SRE), a genetic element that connects cellular stimulation with gene transcription. Elk-1 plays an important role in the regulation of cellular proliferation and apoptosis, thymocyte development, glucose homeostasis and brain function. The biological function of Elk-1 relies essentially on the interaction with other proteins. Elk-1 binds to SRF and generates a functional ternary complex that is required to activate SRE-mediated gene transcription. Elk-1 is kept in an inactive state under basal conditions via binding of a SUMO-histone deacetylase complex. Phosphorylation by extracellular signal-regulated protein kinase, c-Jun N-terminal protein kinase or p38 upregulates the transcriptional activity of Elk-1, mediated by binding to the mediator of RNA polymerase II transcription (Mediator) and the transcriptional coactivator p300. Strong and extended phosphorylation of Elk-1 attenuates Mediator and p300 recruitment and allows the binding of the mSin3A-histone deacetylase corepressor complex. The subsequent dephosphorylation of Elk-1, catalyzed by the protein phosphatase calcineurin, facilitates the re-SUMOylation of Elk-1, transforming Elk-1 back to a transcriptionally inactive state. Thus, numerous protein-protein interactions control the activation cycle of Elk-1 and are essential for its biological function.


Assuntos
Proteínas Elk-1 do Domínio ets/metabolismo , Proteínas Elk-1 do Domínio ets/fisiologia , Animais , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Camundongos , Proteínas Nucleares/metabolismo , Fosforilação , Domínios e Motivos de Interação entre Proteínas/fisiologia , Mapeamento de Interação de Proteínas/métodos , Mapas de Interação de Proteínas/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Fator de Resposta Sérica/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica/genética , Ativação Transcricional/genética , Proteínas Elk-1 do Domínio ets/genética
2.
Pharmacol Res ; 159: 104983, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32504838

RESUMO

A hallmark of diabetes mellitus is the inability of pancreatic ß-cells to secrete sufficient amounts of insulin for maintaining normoglycemia. The formation of smaller islets may underlie the development of a diabetic phenotype, as a decreased ß-cell mass will produce an insufficient amount of insulin. For a pharmacological intervention it is crucial to identify the proteins determining ß-cell mass. Here, we identified the ternary complex factor (TCF) Elk-1 as a regulator of the size of pancreatic islets. Elk-1 mediates, together with a dimer of the serum-response factor (SRF), serum response element-regulated gene transcription. Elk-1 is activated in glucose-treated pancreatic ß-cells but the biological functions of this protein in ß-cells are so far unknown. Elk-1 and homologous TCF proteins are expressed in islets and insulinoma cells. Gene targeting experiments revealed that the TCF proteins show redundant activities. To solve the problem of functional redundancy of these homologous proteins, we generated conditional transgenic mice expressing a dominant-negative mutant of Elk-1 in pancreatic ß-cells. The mutant competes with the wild-type TCFs for DNA and SRF-binding. Expression of the Elk-1 mutant in pancreatic ß-cells resulted in the generation of significantly smaller islets and increased caspase-3 activity, indicating that apoptosis was responsible for the reduction of the pancreatic islet size. Glucose tolerance tests revealed that transgenic mice expressing the dominant-negative mutant of Elk-1 in pancreatic ß-cells displayed impaired glucose tolerance. Thus, we show here for the first time that TCF controls important functions of pancreatic ß-cells in vivo. Elk-1 may be considered as a new therapeutic target for the treatment of diabetes.


Assuntos
Glicemia/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Elk-1 do Domínio ets/metabolismo , Animais , Apoptose , Caspase 3/metabolismo , Linhagem Celular Tumoral , Tamanho Celular , Homeostase , Células Secretoras de Insulina/patologia , Insulinoma/genética , Insulinoma/metabolismo , Insulinoma/patologia , Camundongos Transgênicos , Mutação , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Ratos , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Proteínas Elk-1 do Domínio ets/genética
3.
Gene ; 787: 145645, 2021 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-33848575

RESUMO

Receptors and ion channels expressed on the cell surface ensure proper communication between the cells and the environment. In multicellular organism, stimulus-regulated gene transcription is the basis for communication with the environment allowing individual cells to respond to stimuli such as nutrients, chemical stressors and signaling molecules released by other cells of the organism. Hormones, cytokines, and mitogens bind to receptors and ion channels and induce intracellular signaling cascades involving second messengers, kinases, phosphatases, and changes in the concentration of particular ions. Ultimately, the signaling cascades reach the nucleus. Transcription factors are activated that respond to cellular stimulation and induce changes in gene transcription. Investigating stimulus-transcription coupling combines cell biology with genetics. In this review, we discuss the molecular biology of stimulus-induced transcriptional activators and their responsiveness to extracellular and intracellular signaling molecules and to epigenetic regulators. Stimulus-induced gene expression is measured by several methods, including detection of nuclear translocation of transcription factors, phosphorylation or DNA binding. In this article, we emphasize that the most reliable method to directly measure transcriptional activation involves the use of chromatin-embedded reporter genes.


Assuntos
Cromatina/genética , Genes Reporter , Fatores de Transcrição/metabolismo , Ativação Transcricional , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Regulação da Expressão Gênica , Humanos , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/metabolismo , Transdução de Sinais , Fator de Transcrição AP-1/metabolismo , Proteínas Elk-1 do Domínio ets/metabolismo
4.
Biochem Pharmacol ; 193: 114748, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34461116

RESUMO

Cav1.2 L-type voltage-gated Ca2+ channels play a central role in pancreatic ß-cells by integrating extracellular signals with intracellular signaling events leading to insulin secretion and altered gene transcription. Here, we investigated the intracellular signaling pathway following stimulation of Cav1.2 Ca2+ channels and addressed the function of the transcription factor activator protein-1 (AP-1) in pancreatic ß-cells of transgenic mice. Stimulation of Cav1.2 Ca2+ channels activates AP-1 in insulinoma cells. Pharmacological and genetic experiments identified c-Jun N-terminal protein kinase as a signal transducer connecting Cav1.2 Ca2+ channel activation with gene transcription. Moreover, the basic region-leucine zipper proteins ATF2 and c-Jun or c-Jun-related proteins were involved in stimulus-transcription coupling. We addressed the functions of AP-1 in pancreatic ß-cells analyzing a newly generated transgenic mouse model. These transgenic mice expressed A-Fos, a mutant of c-Fos that attenuates DNA binding of c-Fos dimerization partners. In insulinoma cells, A-Fos completely blocked AP-1 activation following stimulation of Cav1.2 Ca2+ channels. The analysis of transgenic A-Fos-expressing mice revealed that the animals displayed impaired glucose tolerance. Thus, we show here for the first time that AP-1 controls an important function of pancreatic ß-cells in vivo, the regulation of glucose homeostasis.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Fator de Transcrição AP-1/metabolismo , Fator 2 Ativador da Transcrição/genética , Fator 2 Ativador da Transcrição/metabolismo , Animais , Benzamidas/química , Benzamidas/farmacologia , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Linhagem Celular Tumoral , Regulação da Expressão Gênica/fisiologia , Intolerância à Glucose , MAP Quinase Quinase Quinase 1/genética , MAP Quinase Quinase Quinase 1/metabolismo , Camundongos , Camundongos Transgênicos , Piridinas/química , Piridinas/farmacologia , Pirimidinas/química , Pirimidinas/farmacologia , Interferência de RNA , Ratos , Fator de Transcrição AP-1/genética
5.
Metallomics ; 12(11): 1735-1747, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-33030499

RESUMO

Zinc, a trace element, is necessary for the correct structure and function of many proteins. Therefore, Zn2+ has to be taken up by the cells, using specific Zn2+ transporters or Ca2+ channels. In this study, we have focused on two Ca2+ channels, the L-type voltage-gated Cav1.2 channel and the transient receptor potential channel TRPM3. Stimulation of either channel induces an intracellular signaling cascade leading to the activation of the transcription factor AP-1. The influx of Ca2+ ions into the cytoplasm is essential for this activity. We asked whether extracellular Zn2+ ions affect Cav1.2 or TRPM3-induced gene transcription following stimulation of the channels. The results show that extracellular Zn2+ ions reduced the activation of AP-1 by more than 80% following stimulation of either voltage-gated Cav1.2 channels or TRPM3 channels. Experiments performed with cells maintained in Ca2+-free medium revealed that Zn2+ ions cannot replace Ca2+ ions in inducing gene transcription via stimulation of Cav1.2 and TRPM3 channels. Re-addition of Ca2+ ions to the cell culture medium, however, restored the ability of these Ca2+ channels to induce a signaling cascade leading to the activation of AP-1. Secretory cells, including neurons and pancreatic ß-cells, release Zn2+ ions during exocytosis. We propose that the released Zn2+ ions function as a negative feedback loop for stimulus-induced exocytosis by inhibiting Ca2+ channel signaling.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Canais de Cátion TRPM/metabolismo , Transcrição Gênica , Zinco/farmacologia , Animais , Células HEK293 , Humanos , Insulinoma/genética , Íons , Proteína Quinase C/metabolismo , Ratos , Transdução de Sinais , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Transcrição Gênica/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
6.
Biochem Pharmacol ; 170: 113678, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31654626

RESUMO

Transient receptor potential melastatin-8 (TRPM8) channels are activated by cold temperature, menthol and icilin, leading to cold sensation. TRPM8 activation is connected with various diseases, indicating that a specific pharmacological antagonist, allowing nongenetic channel suppression, would be a valuable tool for therapy and basic research. Here, we assessed the biological activity and specificity of various TRPM8 inhibitors following stimulation of TRPM8 channels with either icilin or menthol. Recently, we showed that icilin strikingly upregulates the transcriptional activity of AP-1. By measuring AP-1 activity, we assessed which compound interrupted the TRPM8-induced intracellular signaling cascade from the plasma membrane to the nucleus. We tested the specificity of various TRPM8 inhibitors by analyzing AP-1 activation following stimulation of TRPM3 and TRPV1 channels, L-type voltage-gated Ca2+ channels, and Gαq-coupled receptors, either in the presence or absence of a particular TRPM8 inhibitor. The results show that the TRPM8 inhibitors BCTC, RQ-00203078, TC-1 2014, 2-APB, and clotrimazole blocked TRPM8-mediated activation of AP-1. However, only the compound RQ-00203078 showed TRPM8-specificity, while the other compounds function as broad-spectrum Ca2+ channel inhibitors. In addition, we show that progesterone interfered with TRPM8-induced activation of AP-1, as previously shown for TRPM3 and TRPC6 channels. TRPM8-induced transcriptional activation of AP-1 was additionally blocked by the compound PD98059, indicating that extracellular signal-regulated protein kinase-1/2 is essential to couple TRPM8 stimulation with transcriptional activation of AP-1. Moreover, an influx of Ca2+-ions is essential to induce the intracellular signaling cascade leading to the activation of AP-1.


Assuntos
Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/metabolismo , Transcrição Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Pirazinas/farmacologia , Piridinas/farmacologia , Pirimidinonas/farmacologia , Transcrição Gênica/fisiologia
7.
Int Rev Cell Mol Biol ; 335: 167-189, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29305012

RESUMO

Transient receptor potential (TRP) channels belong to a heterogeneous superfamily of cation channels that are involved in the regulation of numerous biological functions, including regulation of Ca2+ and glucose homeostasis, tumorigenesis, temperature, and pain sensation. To understand the functions of TRP channels, their associated intracellular signaling pathways and molecular targets have to be identified on the cellular level. Stimulation of TRP channels frequently induces an influx of Ca2+ ions into the cells and the subsequent activation of protein kinases. These intracellular signal transduction pathways ultimately induce changes in the gene expression pattern of the cells. Here, we review the effects of TRPC6, TRPM3, and TRPV1 channel stimulation on the activation of the stimulus-responsive transcription factors AP-1, CREB, Egr-1, Elk-1, and NFAT. Following activation, these transcription factors induce the transcription of delayed response genes. We propose that many biological functions of TRP channels can be explained by the activation of stimulus-responsive transcription factors and their delayed response genes. The proteins encoded by those delayed response genes may be responsible for the biochemical and physiological changes following TRP channel activation.


Assuntos
Transcrição Gênica , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Sequência de Bases , Humanos , Modelos Biológicos , Fatores de Transcrição/metabolismo
8.
Biochem Pharmacol ; 150: 160-169, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29452097

RESUMO

Transient receptor potential vanilloid 1 (TRPV1) channels were originally described as the receptors of capsaicin, the main constituent of hot chili pepper. The biological functions of TRPV1 channels include pain sensation and inflammatory thermal hyperalgesia. Here, we show that stimulation of HEK293 cells expressing TRPV1 channels (H2C1 cells) with capsaicin or the TRPV1 ligand resiniferatoxin activated transcription mediated by the transcription factor AP-1. No cell death was occurring under these experimental conditions. The AP-1 activity was not altered in capsaicin or resiniferatoxin-stimulated HEK293 cells lacking TRPV1. We identified the AP-1 DNA binding site as the capsaicin/resiniferatoxin-responsive element. Stimulation with the TRPV1 ligand N-arachidonoyldopamine increased AP-1 activity in a TRPV1-dependent and TRPV1-independent manner. Stimulation of TRPV1 channels induced an influx of Ca2+ into the cells and this rise in intracellular Ca2+ was essential for activating AP-1 in capsaicin or resiniferatoxin-stimulated cells. N-arachidonoyldopamine stimulation induced a rise in intracellular Ca2+ in a TRPV-1 dependent and independent manner. AP-1 is a dimeric transcription factor, composed of proteins of the c-Jun, c-Fos and ATF families. Stimulation of TRPV1 channels with capsaicin increased c-Jun and c-Fos biosynthesis in H2C1 cells. The signal transduction of capsaicin, leading to enhanced AP-1-mediated transcription, required extracellular signal-regulated protein kinase ERK1/2 as a signal transducer and the activation of the transcription factors c-Jun and ternary complex factor. Together, these data suggest that the intracellular functions of TRPV1 stimulation may rely on the activation of a stimulus-regulated protein kinase and stimulus-responsive transcription factors.


Assuntos
Capsaicina/farmacologia , Canais de Cátion TRPV/metabolismo , Fator de Transcrição AP-1/metabolismo , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Fármacos do Sistema Sensorial/farmacologia , Canais de Cátion TRPV/agonistas , Fator de Transcrição AP-1/agonistas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA