Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Mol Cell Cardiol ; 127: 31-43, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30521840

RESUMO

The sympathetic nervous system is the main stimulator of cardiac function. While acute activation of the ß-adrenoceptors exerts positive inotropic and lusitropic effects by increasing cAMP and Ca2+, chronically enhanced sympathetic tone with changed ß-adrenergic signaling leads to alterations of gene expression and remodeling. The CREB-regulated transcription coactivator 1 (CRTC1) is activated by cAMP and Ca2+. In the present study, the regulation of CRTC1 in cardiomyocytes and its effect on cardiac function and growth was investigated. In cardiomyocytes, isoprenaline induced dephosphorylation, and thus activation of CRTC1, which was prevented by propranolol. Crtc1-deficient mice exhibited left ventricular dysfunction, hypertrophy and enlarged cardiomyocytes. However, isoprenaline-induced contractility of isolated trabeculae or phosphorylation of cardiac troponin I, cardiac myosin-binding protein C, phospholamban, and ryanodine receptor were not altered, suggesting that cardiac dysfunction was due to the global lack of Crtc1. The mRNA and protein levels of the Gαq GTPase activating protein regulator of G-protein signaling 2 (RGS2) were lower in hearts of Crtc1-deficient mice. Chromatin immunoprecipitation and reporter gene assays showed stimulation of the Rgs2 promoter by CRTC1. In Crtc1-deficient cardiomyocytes, phosphorylation of the Gαq-downstream kinase ERK was enhanced. CRTC1 content was higher in cardiac tissue from patients with aortic stenosis or hypertrophic cardiomyopathy and from two murine models mimicking these diseases. These data suggest that increased CRTC1 in maladaptive hypertrophy presents a compensatory mechanism to delay disease progression in part by enhancing Rgs2 gene transcription. Furthermore, the present study demonstrates an important role of CRTC1 in the regulation of cardiac function and growth.


Assuntos
Cardiomegalia/metabolismo , Fatores de Transcrição/metabolismo , Animais , Cardiomegalia/diagnóstico por imagem , Cardiomegalia/fisiopatologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Proteínas RGS/genética , Proteínas RGS/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais , Fatores de Transcrição/deficiência
2.
Arch Pharm (Weinheim) ; 349(6): 410-3, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27100796

RESUMO

Diabetes mellitus is one of the most rapidly increasing diseases worldwide, whereby approximately 90-95% of patients suffer from type 2 diabetes. Considering its micro- and macrovascular complications like blindness and myocardial infarction, a reliable anti-diabetic treatment is needed. Maintaining the function and the mass of the insulin producing beta-cells despite elevated levels of beta-cell-toxic prediabetic signals represents a desirable mechanism of action of anti-diabetic drugs. The dual leucine zipper kinase (DLK) inhibits the action of two transcription factors within the beta-cell, thereby interfering with insulin secretion and production and the conservation of beta-cell mass. Furthermore, DLK action is regulated by prediabetic signals. Hence, the inhibition of this kinase might protect beta-cells against beta-cell-toxic prediabetic signals and prevent the development of diabetes. DLK might thus present a novel drug target for the treatment of diabetes mellitus type 2.


Assuntos
Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/enzimologia , MAP Quinase Quinase Quinases/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/enzimologia , Humanos , MAP Quinase Quinase Quinases/antagonistas & inibidores , Terapia de Alvo Molecular/métodos
3.
Cells ; 13(4)2024 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-38391946

RESUMO

The dual leucine zipper kinase (DLK) alias mitogen-activated protein 3 kinase 12 (MAP3K12) has gained much attention in recent years. DLK belongs to the mixed lineage kinases, characterized by homology to serine/threonine and tyrosine kinase, but exerts serine/threonine kinase activity. DLK has been implicated in many diseases, including several neurodegenerative diseases, glaucoma, and diabetes mellitus. As a MAP3K, it is generally assumed that DLK becomes phosphorylated and activated by upstream signals and phosphorylates and activates itself, the downstream serine/threonine MAP2K, and, ultimately, MAPK. In addition, other mechanisms such as protein-protein interactions, proteasomal degradation, dephosphorylation by various phosphatases, palmitoylation, and subcellular localization have been shown to be involved in the regulation of DLK activity or its fine-tuning. In the present review, the diverse mechanisms regulating DLK activity will be summarized to provide better insights into DLK action and, possibly, new targets to modulate DLK function.


Assuntos
Zíper de Leucina , MAP Quinase Quinase Quinases , MAP Quinase Quinase Quinases/metabolismo , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Serina/metabolismo , Treonina/metabolismo
4.
Naunyn Schmiedebergs Arch Pharmacol ; 396(6): 1223-1233, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36700987

RESUMO

The dual leucine zipper kinase (DLK) and the ubiquitously expressed transcription factor c-FOS have important roles in beta-cell proliferation and function. Some studies in neuronal cells suggest that DLK can influence c-FOS expression. Given that c-FOS is mainly regulated at the transcriptional level, the effect of DLK on c-FOS promoter activity was investigated in the beta-cell line HIT. The methods used in this study are the following: Luciferase reporter gene assays, immunoblot analysis, CRISPR-Cas9-mediated genome editing, and real-time quantitative PCR. In the beta-cell line HIT, overexpressed DLK increased c-FOS promoter activity twofold. Using 5'-,3'-promoter deletions, the promoter regions from - 348 to - 339 base pairs (bp) and from a - 284 to - 53 bp conferred basal activity, whereas the promoter region from - 711 to - 348 bp and from - 53 to + 48 bp mediated DLK responsiveness. Mutation of the cAMP response element within the promoter prevented the stimulatory effect of DLK. Treatment of HIT cells with KCl and the adenylate cyclase activator forskolin increased c-FOS promoter transcriptional activity ninefold. Since the transcriptional activity of those promoter fragments activated by KCl and forskolin was decreased by DLK, DLK might interfere with KCl/forskolin-induced signaling. In a newly generated, genome-edited HIT cell line lacking catalytically active DLK, c-Fos mRNA levels were reduced by 80% compared to the wild-type cell line. DLK increased c-FOS promoter activity but decreased stimulated transcriptional activity, suggesting that DLK fine-tunes c-FOS promoter-dependent gene transcription. Moreover, at least in HIT cells, DLK is required for FOS mRNA expression.


Assuntos
Zíper de Leucina , MAP Quinase Quinase Quinases , Colforsina , MAP Quinase Quinase Quinases/metabolismo , Linhagem Celular , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo
5.
Exp Dermatol ; 20(3): 232-6, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21323745

RESUMO

Unlike other immunosuppressive drugs including everolimus, cyclosporin A causes a dramatic increase of UV-induced skin cancer, a feature that is reminiscent of xeroderma pigmentosum (XP), where defective nucleotide excision repair (NER) of UV-induced DNA damage results in cutaneous carcinogenesis. The molecular basis of the clinically important differential activities of cyclosporin A and everolimus is still unclear. We measured post-UV cell survival of cyclosporin A- and everolimus-treated human fibroblasts and lymphoblasts using a cell proliferation assay (MTT). The cellular NER capacity was assessed by host cell reactivation. Using an ELISA and specific antibodies, cyclobutane pyrimidine and pyrimidine-6,4-pyrimidone photoproduct removal from the cellular genome was measured. The effect of calcineurin on NER was investigated using a calcineurin A expression vector and specific RNAi. Cyclosporin A led to a dose dependent decrease in post-UV cell survival, inhibited NER and blocked photoproduct removal. In contrast, none of these effects where seen in everolimus-treated cells. Overexpression of calcineurin A resulted in increased NER and complemented the Cyclosporin A-induced reduction of NER. Downregulation of calcineurin using RNAi inhibited NER comparable to cyclosporin A-treatment. We conclude that cyclosporin A, but not everolimus, leads to an increased skin cancer risk via a calcineurin signalling-dependent impairment of NER.


Assuntos
Calcineurina/metabolismo , Ciclosporina/farmacologia , Reparo do DNA/efeitos dos fármacos , Terapia de Imunossupressão/efeitos adversos , Neoplasias/etiologia , Sirolimo/análogos & derivados , Calcineurina/genética , Linhagem Celular Transformada , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Ciclosporina/efeitos adversos , Everolimo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Humanos , Imunossupressores/farmacologia , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Linfócitos/efeitos da radiação , Fosforilação/efeitos dos fármacos , Dímeros de Pirimidina/metabolismo , RNA Interferente Pequeno/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Sirolimo/farmacologia , Transfecção , Raios Ultravioleta
6.
Cell Signal ; 82: 109953, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33600948

RESUMO

Hyperglycemia enhancing the intracellular levels of reactive oxygen species (ROS) contributes to dysfunction and progressive loss of beta cells and thereby to diabetes mellitus. The oxidation sensitive calcium/calmodulin dependent phosphatase calcineurin promotes pancreatic beta cell function and survival whereas the dual leucine zipper kinase (DLK) induces apoptosis. Therefore, it was studied whether calcineurin interferes with DLK action. In a beta cell line similar concentrations of H2O2 decreased calcineurin activity and activated DLK. DLK interacted via its φLxVP motif (aa 362-365) with the interface of the calcineurin subunits A and B. Mutation of the Val prevented this protein protein interaction, hinting at a distinct φLxVP motif. Indeed, mutational analysis revealed an ordered structure of DLK's φLxVP motif whereby Val mediates the interaction with calcineurin and Leu maintains an enzymatically active conformation. Overexpression of DLK wild-type but not the DLK mutant unable to bind calcineurin diminished calcineurin-induced nuclear localisation of the nuclear factor of activated T-cells (NFAT), suggesting that both, DLK and NFAT compete for the substrate binding site of calcineurin. The calcineurin binding-deficient DLK mutant exhibited increased DLK activity measured as phosphorylation of the downstream c-Jun N-terminal kinase, inhibition of CRE-dependent gene transcription and induction of apoptosis. These findings show that calcineurin interacts with DLK; and inhibition of calcineurin increases DLK activity. Hence, this study demonstrates a novel mechanism regulating DLK action. These findings suggest that ROS through inhibition of calcineurin enhance DLK activity and thereby lead to beta cell dysfunction and loss and ultimately diabetes mellitus.


Assuntos
Calcineurina/metabolismo , Células Secretoras de Insulina , MAP Quinase Quinase Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Apoptose , Linhagem Celular , Cricetinae , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Ligação Proteica
7.
Biosci Rep ; 29(2): 77-87, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18717645

RESUMO

Lithium salts are clinically important drugs used to treat bipolar mood disorder. The mechanisms accounting for the clinical efficacy are not completely understood. Chronic treatment with lithium is required to establish mood stabilization, suggesting the involvement of neuronal plasticity processes. CREB (cAMP-response-element-binding protein) is a transcription factor known to mediate neuronal adaptation. Recently, the CREB-co-activator TORC (transducer of regulated CREB) has been identified as a novel target of lithium and shown to confer an enhancement of cAMP-induced CREB-directed gene transcription by lithium. TORC is sequestered in the cytoplasm and its nuclear translocation controls CREB activity. In the present study, the effect of lithium on TORC function was investigated. Lithium affected neither the nuclear translocation of TORC nor TORC1 transcriptional activity, but increased the promoter occupancy by TORC1 as revealed by chromatin immunoprecipitation assay. In a mammalian two-hybrid assay, as well as in a cell-free GST (glutathione transferase) pull-down assay, lithium enhanced the CREB-TORC1 interaction. Magnesium ions strongly inhibited the interaction between GST-CREB and TORC1 and this effect was reversed by lithium. Thus our results suggest that, once TORC has entered the nucleus, lithium as a cation stimulates directly the binding of TORC to CREB, leading to an increase in cAMP-induced CREB target-gene transcription. This novel mechanism of lithium action is likely to contribute to the clinical mood-stabilizing effect of lithium salts.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Lítio/farmacologia , Fatores de Transcrição/metabolismo , Ativação Transcricional/efeitos dos fármacos , Animais , Núcleo Celular/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina , Cricetinae , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Glutationa Transferase/metabolismo , Imuno-Histoquímica , Insulinoma/metabolismo , Insulinoma/patologia , Modelos Biológicos , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição/genética , Transfecção , Técnicas do Sistema de Duplo-Híbrido
8.
Mol Pharmacol ; 73(3): 652-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18042735

RESUMO

Post-transplant diabetes is an untoward effect often observed under immunosuppressive therapy with cyclosporin A. Besides the development of peripheral insulin resistance and a decrease in insulin gene transcription, a beta-cell toxic effect has been described. However, its molecular mechanism remains unknown. In the present study, the effect of cyclosporin A and the dual leucine-zipper-bearing kinase (DLK) on beta-cell survival was investigated. Cyclosporin A decreased the viability of the insulin-producing pancreatic islet cell line HIT in a time- and concentration-dependent manner. Upon exposure to the immunosuppressant fragmentation of DNA, the activation of the effector caspase-3 and a decrease of full-length caspase-3 and Bcl(XL) were observed in HIT cells and in primary mature murine islets, respectively. Cyclosporin A and tacrolimus, both potent inhibitors of the calcium/calmodulin-dependent phosphatase calcineurin, stimulated the enzymatic activity of cellular DLK in an in vitro kinase assay. Immunocytochemistry revealed that the overexpression of DLK but not its kinase-dead mutant induced apoptosis and enhanced cyclosporin A-induced apoptosis to a higher extent than the drug alone. Moreover, in the presence of DLK, the effective concentration for cyclosporin A-caused apoptosis was similar to its known IC(50) value for the inhibition of calcineurin activity in beta cells. These data suggest that cyclosporin A through inhibition of calcineurin activates DLK, thereby leading to beta-cell apoptosis. This action may thus be a novel mechanism through which cyclosporin A precipitates post-transplant diabetes.


Assuntos
Apoptose/efeitos dos fármacos , Ciclosporina/farmacologia , Imunossupressores/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , MAP Quinase Quinase Quinases/metabolismo , Animais , Inibidores de Calcineurina , Caspase 3/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Fragmentação do DNA , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Formazans/metabolismo , Imuno-Histoquímica , Concentração Inibidora 50 , Cinética , MAP Quinase Quinase Quinases/análise , Camundongos , Estatística como Assunto , Tacrolimo/farmacologia , Sais de Tetrazólio/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
9.
Mol Pharmacol ; 73(2): 509-17, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17962386

RESUMO

The peptide hormone glucagon stimulates hepatic glucose output, and its levels in the blood are elevated in type 2 diabetes mellitus. The nuclear receptor peroxisome proliferator-activated receptor-gamma (PPARgamma) has essential roles in glucose homeostasis, and thiazolidinedione PPARgamma agonists are clinically important antidiabetic drugs. As part of their antidiabetic effect, thiazolidinediones such as rosiglitazone have been shown to inhibit glucagon gene transcription through binding to PPARgamma and inhibition of the transcriptional activity of PAX6 that is required for cell-specific activation of the glucagon gene. However, how thiazolidinediones and PPARgamma inhibit PAX6 activity at the glucagon promoter remained unknown. After transient transfection of a glucagon promoter-reporter fusion gene into a glucagon-producing pancreatic islet alpha-cell line, ligand-bound PPARgamma was found in the present study to inhibit glucagon gene transcription also after deletion of its DNA-binding domain. Like PPARgamma ligands, also retinoid X receptor (RXR) agonists inhibited glucagon gene transcription in a PPARgamma-dependent manner. In glutathione transferase pull-down assays, the ligand-bound PPARgamma-RXR heterodimer bound to the transactivation domain of PAX6. This interaction depended on the presence of the ligand and RXR, but it was independent of the PPARgamma DNA-binding domain. Chromatin immunoprecipitation experiments showed that PPARgamma is recruited to the PAX6-binding proximal glucagon promoter. Taken together, the results of the present study support a model in which a ligand-bound PPARgamma-RXR heterodimer physically interacts with promoter-bound PAX6 to inhibit glucagon gene transcription. These data define PAX6 as a novel physical target of PPARgamma-RXR.


Assuntos
Proteínas do Olho/metabolismo , Glucagon/antagonistas & inibidores , Glucagon/genética , Proteínas de Homeodomínio/metabolismo , PPAR gama/metabolismo , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/metabolismo , Receptor X Retinoide gama/metabolismo , Transcrição Gênica/fisiologia , Ativação Transcricional/fisiologia , Animais , Linhagem Celular , Dimerização , Proteínas do Olho/genética , Proteínas do Olho/fisiologia , Glucagon/biossíntese , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Fator de Transcrição PAX6 , PPAR gama/genética , PPAR gama/fisiologia , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/fisiologia , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia , Receptor X Retinoide gama/genética
10.
Gene ; 386(1-2): 52-62, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17010535

RESUMO

The serotonin receptor type 3 is a pentameric ligand-gated ion channel regulating intestinal motility, nausea, and vomiting in humans. The HTR3B gene codes for the subunit B of this receptor. The HTR3B transcription start site is not unequivocally identified. In the present study we used transcription start site analyses, transcript-specific RT-PCR, and functional promoter analyses to identify the 5' structure of the HTR3B gene. According to these experiments, two alternative promoters control the expression of different HTR3B transcripts in the peripheral and central nervous system. The transcription start sites observed in the intestine corresponded to the current human genome annotation (NCBI Build 36.1, March 2006). The transcription start sites in the brain, however, were localized in a region about 4000 bp downstream. The brain transcripts lacked the coding first exon of the HTR3B structure published earlier but had an upstream-extended exon 2 containing a new potential translational start site. Reporter gene analyses showed significant promoter activity of the genomic region located 1560 bp upstream to 93 bp downstream of the brain-specific transcription start sites. This data suggests a different transcriptional regulation of the HTR3B gene in the peripheral and the central nervous system that leads to the expression of transcripts with variations in the 5' coding sequence. Further studies on the expression, structure and function of therefore expected tissue-specific 5-HT(3B) isoforms are required.


Assuntos
Processamento Alternativo , Encéfalo/metabolismo , Intestino Delgado/metabolismo , Regiões Promotoras Genéticas , Receptores de Serotonina/genética , Animais , Sequência de Bases , Humanos , Dados de Sequência Molecular , Especificidade de Órgãos/genética , Células PC12 , Sistema Nervoso Periférico/metabolismo , Subunidades Proteicas/biossíntese , Subunidades Proteicas/genética , Ratos , Receptores de Serotonina/biossíntese , Receptores 5-HT3 de Serotonina
11.
Naunyn Schmiedebergs Arch Pharmacol ; 390(8): 813-825, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28550372

RESUMO

Reduction in beta-cell mass and function contributes to the pathogenesis of diabetes mellitus type 2. The proinflammatory cytokines tumor necrosis factor (TNF)α and interleukin (IL)-1ß have been implicated in the pathogenesis of this disease. Overexpression of the dual leucine zipper kinase (DLK) inhibits beta-cell function and induces apoptosis in the beta-cell line HIT. In the present study, it was investigated whether TNFα or IL-1ß stimulates DLK enzymatic activity. Immunoblot analysis, transient transfection with luciferase reporter gene assays, and immunofluorescence were used. In contrast to IL-1ß, TNFα stimulated DLK kinase activity, which was dependent on the c-Jun N-terminal kinase (JNK). Furthermore, DLK contributed to TNFα-induced JNK phosphorylation. The phosphorylation of DLK on Ser-302 within the activation loop was required for DLK to stimulate JNK and to inhibit CREB-dependent gene transcription. TNFα induced apoptosis in a time- and concentration-dependent manner and inhibited CREB-directed gene transcription in HIT cells. The reduction of endogenous DLK by small interfering or small hairpin RNA attenuated TNFα's effects on apoptosis and CREB-dependent transcription. These data suggest that TNFα induces beta-cell apoptosis through activation of DLK thereby inhibiting the beta-cell protective transcription factor CREB. Furthermore, activation of DLK by a well-known diabetic risk factor supports the role of DLK in the pathogenesis of diabetes mellitus. Thus, the inhibition of DLK might prevent or retard the pathogenesis of diabetes mellitus type 2.


Assuntos
Células Secretoras de Insulina/efeitos dos fármacos , MAP Quinase Quinase Quinases/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Cricetinae , Células Secretoras de Insulina/metabolismo , Interleucina-1beta/farmacologia
12.
Mol Cell Endocrinol ; 441: 108-115, 2017 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-27889473

RESUMO

Endothelial cell-derived products influence the synthesis of aldosterone and cortisol in human adrenocortical cells by modulating proteins such as steroidogenic acute-regulatory (StAR) protein, steroidogenic factor (SF)-1 and CITED2. However, the potential endothelial cell-derived factors that mediate this effect are still unknown. The current study was perfomed to look into the control of ß-catenin activity by endothelial cell-derived factors and to identify a mechanism by which they affect ß-catenin activity in adrenocortical NCIH295R cells. Using reporter gene assays and Western blotting, we found that endothelial cell-conditioned medium (ECCM) led to nuclear translocation of ß-catenin and an increase in ß-catenin-dependent transcription that could be blocked by U0126, an inhibitor of the mitogen-activated protein kinase pathway. Furthermore, we found that a receptor tyrosin kinase (RTK) was involved in ECCM-induced ß-catenin-dependent transcription. Through selective inhibition of RTK using Su5402, it was shown that receptors responding to basic fibroblast growth factor (bFGF) mediate the action of ECCM. Adrenocortical cells treated with bFGF showed a significant greater level of bFGF mRNA. In addition, HUVECs secrete bFGF in a density-dependent manner. In conclusion, the data suggest that endothelial cells regulate ß-catenin activity in adrenocortical cells also via secretion of basic fibroblast growth factor.


Assuntos
Córtex Suprarrenal/citologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , beta Catenina/metabolismo , Linhagem Celular , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Luciferases/metabolismo , Proteínas Quinases/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos
13.
Biochem J ; 389(Pt 3): 831-41, 2005 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-15828872

RESUMO

The pancreatic islet hormone glucagon stimulates hepatic glucose production and thus maintains blood glucose levels in the fasting state. Transcription factors of the Foxa [Fox (forkhead box) subclass A; also known as HNF-3 (hepatocyte nuclear factor-3)] family are required for cell-specific activation of the glucagon gene in pancreatic islet alpha-cells. However, their action on the glucagon gene is poorly understood. In the present study, comparative sequence analysis and molecular characterization using protein-DNA binding and transient transfection assays revealed that the well-characterized Foxa-binding site in the G2 enhancer element of the rat glucagon gene is not conserved in humans and that the human G2 sequence lacks basal enhancer activity. A novel Foxa site was identified that is conserved in rats, mice and humans. It mediates activation of the glucagon gene by Foxa proteins and confers cell-specific promoter activity in glucagon-producing pancreatic islet alpha-cell lines. In contrast with previously identified Foxa-binding sites in the glucagon promoter, which bind nuclear Foxa2, the novel Foxa site was found to bind preferentially Foxa1 in nuclear extracts of a glucagon-producing pancreatic islet alpha-cell line, offering a mechanism that explains the decrease in glucagon gene expression in Foxa1-deficient mice. This site is located just upstream of the TATA box (between -30 and -50), suggesting a role for Foxa proteins in addition to direct transcriptional activation, such as a role in opening the chromatin at the start site of transcription of the glucagon gene.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Glucagon/genética , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Células Cultivadas , Sequência Conservada , Regulação da Expressão Gênica , Fator 3-alfa Nuclear de Hepatócito , Humanos , Camundongos , Dados de Sequência Molecular , Mutação , Regiões Promotoras Genéticas/fisiologia , Ratos , Homologia de Sequência do Ácido Nucleico
14.
Expert Opin Ther Pat ; 26(5): 607-16, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27043251

RESUMO

INTRODUCTION: The dual leucine zipper kinase (DLK, MAP3K12) is essential for neuronal development and has been shown to mediate axon regeneration. On the other hand, DLK is involved in the pathogenesis of neurodegenerative disease and diabetes mellitus. Several patents have been published claiming to modulate or inhibit DLK by various approaches including ATP competitive inhibitors. In addition, two publications describe SAR of highly selective DLK inhibitors with efficacy in distinct mouse models of neurodegeneration. AREAS COVERED: This review summarized patents claiming to modulate DLK activity published between 2010 and 2015. Peer-reviewed publications related to the patents and additional peer-reviewed publications are included. This article describes 18 patents from three pharmaceutical companies and three academic research groups. EXPERT OPINION: Several methods are proposed to modulate DLK activity, some of them very experimental and not suitable for easy application in patients. ATP competitive kinase inhibitors exert high affinity, but for the majority, no information about their selectivity is available. To date, two inhibitors have been tested in mice. Given the controversial findings that DLK is required for neurodegeneration and for axon regeneration, more research is needed to further elucidate the regulation and the function of this kinase in diverse organs/tissues and under physiological and pathological conditions.


Assuntos
Desenho de Fármacos , MAP Quinase Quinase Quinases/efeitos dos fármacos , Doenças Neurodegenerativas/tratamento farmacológico , Animais , Axônios/metabolismo , Modelos Animais de Doenças , Humanos , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Regeneração Nervosa/fisiologia , Doenças Neurodegenerativas/fisiopatologia , Patentes como Assunto
15.
Cell Signal ; 28(4): 272-83, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26776303

RESUMO

The dual leucine zipper kinase DLK induces ß-cell apoptosis by inhibiting the transcriptional activity conferred by the ß-cell protective transcription factor cAMP response element binding protein CREB. This action might contribute to ß-cell loss and ultimately diabetes. Within its kinase domain DLK shares high homology with the mixed lineage kinase (MLK) 3, which is activated by tumor necrosis factor (TNF) α and interleukin (IL)-1ß, known prediabetic signals. In the present study, the regulation of DLK in ß-cells by these cytokines was investigated. Both, TNFα and IL-1ß induced the nuclear translocation of DLK. Mutations within a putative nuclear localization signal (NLS) prevented basal and cytokine-induced nuclear localization of DLK and binding to the importin receptor importin α, thereby demonstrating a functional NLS within DLK. DLK NLS mutants were catalytically active as they phosphorylated their down-stream kinase c-Jun N-terminal kinase to the same extent as DLK wild-type but did neither inhibit CREB-dependent gene transcription nor transcription conferred by the promoter of the anti-apoptotic protein BCL-xL. In addition, the ß-cell apoptosis-inducing effect of DLK was severely diminished by mutation of its NLS. In a murine model of prediabetes, enhanced nuclear DLK was found. These data demonstrate that DLK exerts distinct functions, depending on its subcellular localization and thus provide a novel level of regulating DLK action. Furthermore, the prevention of the nuclear localization of DLK as induced by prediabetic signals with consecutive suppression of ß-cell apoptosis might constitute a novel target in the therapy of diabetes mellitus.


Assuntos
Apoptose , Núcleo Celular/enzimologia , Diabetes Mellitus Experimental/enzimologia , Células Secretoras de Insulina/enzimologia , MAP Quinase Quinase Quinases/metabolismo , Animais , Linhagem Celular , Núcleo Celular/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/genética , Células Secretoras de Insulina/patologia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase Quinases/genética , Camundongos , Mutação , Transporte Proteico/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
16.
Br J Pharmacol ; 144(7): 982-93, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15711594

RESUMO

Cyclosporin A and tacrolimus are clinically important immunosuppressive drugs directly targeting the transcription factor nuclear factor of activated T cells (NFAT). Through inhibition of calcineurin phosphatase activity they block the dephosphorylation and thus activation of NFAT. Cyclosporin A and tacrolimus also inhibit other calcineurin-dependent transcription factors including the ubiquitously expressed cAMP response element-binding protein (CREB). Membrane depolarization by phosphorylating CREB on Ser119 leads to the recruitment of its coactivator CREB-binding protein (CBP) that stimulates initiation of transcription. It was unknown at what step in CREB-mediated transcription cyclosporin A and tacrolimus interfere. In transient transfection experiments, using GAL4-CREB fusion proteins and a pancreatic islet beta-cell line, cyclosporin A inhibited depolarization-induced activation of CREB proteins which carried various deletions or mutations throughout their sequence providing no evidence for the existence of a distinct CREB domain conferring cyclosporin A sensitivity. In a mammalian two-hybrid assay, cyclosporin A did not inhibit Ser119-dependent interaction of CREB with its coactivator CBP. Using GAL4-CBP fusion proteins, cyclosporin A inhibited depolarization-induced CBP activity, with cyclosporin A-sensitive domains mapped to both the N- (aa 1-451) and C-terminal (aa 2040-2305) ends of CBP. The depolarization-induced transcriptional activity of the CBP C-terminus was enhanced by overexpression of calcineurin and was inhibited by cyclosporin A and tacrolimus in a concentration-dependent manner with IC50 values (10 and 1 nM, respectively) consistent with their known IC50 values for inhibition of calcineurin. These data suggest that, in contrast to NFAT, cyclosporin A and tacrolimus inhibit CREB transcriptional activity at the coactivator level.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/biossíntese , Ciclosporina/farmacologia , Imunossupressores/farmacologia , Proteínas Nucleares/biossíntese , Tacrolimo/farmacologia , Transativadores/biossíntese , Animais , Proteína de Ligação a CREB , Cricetinae , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Proteínas Nucleares/genética , Transativadores/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia
17.
Naunyn Schmiedebergs Arch Pharmacol ; 367(3): 227-36, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12644894

RESUMO

Cyclosporin A and tacrolimus are important immunosuppressive drugs. They share a diabetogenic action as one of their most serious adverse effects. In a single study, tacrolimus (100 nM) inhibited human insulin gene transcription in the beta-cell line HIT. Using transfections of a human insulin-reporter gene into HIT cells, the present study shows that this inhibition is seen only at high concentrations of tacrolimus and is not caused by cyclosporin A. However, after stimulation by the major second messengers in the regulation of the insulin gene, cAMP and depolarization-induced calcium influx, both tacrolimus and cyclosporin A inhibited human insulin gene transcription in a concentration-dependent manner with IC(50) values of 1 nM and 30 nM, respectively. A further analysis offers a mechanism for this effect by revealing that the activation by cAMP and calcium of human insulin gene transcription is mediated by the transcription factor cAMP-responsive element binding protein (CREB) whose activity is inhibited by the immunosuppressants. These data demonstrate for the first time that cAMP- and calcium-induced activity of the human insulin gene is mediated by CREB and blocked by both tacrolimus and cyclosporin A at concentrations that inhibit calcineurin phosphatase activity. Since also the immunosuppressive effects of cyclosporin A and tacrolimus are thought to be secondary to inhibition of calcineurin, the present study suggests that inhibition of human insulin gene transcription by the immunosuppressants is clinically important and may contribute to their diabetogenic effect.


Assuntos
Inibidores de Calcineurina , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Ciclosporina/farmacologia , Insulina/biossíntese , Tacrolimo/farmacologia , Cálcio/metabolismo , Células Cultivadas , AMP Cíclico/metabolismo , Ciclosporina/administração & dosagem , Regulação da Expressão Gênica , Humanos , Imunossupressores/farmacologia , Insulina/genética , Tacrolimo/administração & dosagem , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos
18.
Cell Signal ; 26(9): 1792-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24726898

RESUMO

Insulin biosynthesis is an essential ß-cell function and inappropriate insulin secretion and biosynthesis contribute to the pathogenesis of diabetes mellitus type 2. Previous studies showed that the dual leucine zipper kinase (DLK) induces ß-cell apoptosis. Since ß-cell dysfunction precedes ß-cell loss, in the present study the effect of DLK on insulin gene transcription was investigated in the HIT-T15 ß-cell line. Downregulation of endogenous DLK increased whereas overexpression of DLK decreased human insulin gene transcription. 5'- and 3'-deletion human insulin promoter analyses resulted in the identification of a DLK responsive element that mapped to the DNA binding-site for the ß-cell specific transcription factor MafA. Overexpression of DLK wild-type but not its kinase-dead mutant inhibited MafA transcriptional activity conferred by its transactivation domain. Furthermore, in the non-ß-cell line JEG DLK inhibited MafA overexpression-induced human insulin promoter activity. Overexpression of MafA and DLK or its kinase-dead mutant into JEG cells revealed that DLK but not its mutant reduced MafA protein content. Inhibition of the down-stream DLK kinase c-Jun N-terminal kinase (JNK) by SP600125 attenuated DLK-induced MafA loss. Furthermore, mutation of the serine 65 to alanine, shown to confer MafA protein stability, increased MafA-dependent insulin gene transcription and prevented DLK-induced MafA loss in JEG cells. These data suggest that DLK by activating JNK triggers the phosphorylation and degradation of MafA thereby attenuating insulin gene transcription. Given the importance of MafA for ß-cell function, the inhibition of DLK might preserve ß-cell function and ultimately retard the development of diabetes mellitus type 2.


Assuntos
Regulação da Expressão Gênica , Insulina/genética , Insulina/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Fatores de Transcrição Maf Maior/genética , Fatores de Transcrição Maf Maior/metabolismo , Antracenos/farmacologia , Linhagem Celular , Células HEK293 , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/genética , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas , Interferência de RNA
19.
Cell Signal ; 25(1): 113-25, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23000340

RESUMO

Lithium salts are important drugs to treat bipolar disorder. Previous work showed that lithium by enforcing the interaction between the transcription factor CREB and its coactivator CRTC1 enhanced cAMP-stimulated CREB-dependent gene transcription. Both CREB and CRTC have been implicated in neuronal adaptation, which might underlie lithium's therapeutic action. In the present study the mechanisms of lithium action on cAMP-induced CREB-dependent gene transcription were further elucidated. Transient transfection assays revealed that all three CRTC isoforms conferred lithium responsiveness to CREB whereas their intrinsic transcriptional activities remained unchanged by lithium, suggesting a conformational change of CREB or CRTC by lithium. In in vitro protein-protein interaction assays lithium enhanced the interaction between CREB and both coactivators CRTC and CBP. Furthermore, lithium enforced the oligomerization of CRTC, a prerequisite for CREB interaction. For further evaluation it was investigated whether lithium competes with magnesium, which coordinates the conformation of the CREB basic region leucine zipper (bZip). Mutational analysis of the magnesium coordinating lysine-290 within the bZip, in vitro and intracellular interaction assays and luciferase reporter-gene assays revealed that the effect of lithium on the CREB-CRTC interaction or on the transcriptional activity, respectively, was not affected by the mutation, thus excluding a magnesium-lithium competition. However, the CREB-CRTC interaction was strongly increased in lysine-290-mutants thereby extending the CRTC-CREB interaction domain. Taken together the results exclude a competition between lithium and magnesium at the bZip, but suggest that lithium by enforcing the CRTC-oligomer formation and the interaction of CREB-CBP-CRTC enhances cAMP-induced CREB-dependent gene transcription.


Assuntos
Proteína de Ligação a CREB/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Cloreto de Lítio/farmacologia , Multimerização Proteica/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular Tumoral , Cricetinae , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Genes Reporter , Humanos , Cloreto de Magnésio/farmacologia , Mutação , Mapeamento de Interação de Proteínas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Transfecção
20.
Cell Signal ; 23(2): 344-53, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20940047

RESUMO

CREB is a ubiquitously expressed transcription factor regulating gene expression via binding to a CRE DNA element. Previous work showed that the dual leucine zipper kinase (DLK) reduced CREB-dependent gene transcription at least in part via inhibition of the coactivator CBP. Here we demonstrate that DLK also inhibits CREB activity by affecting the interaction of CREB with its second coactivator TORC. DLK acted on TORC-dependent transcription by distinct mechanisms. An interaction between DLK and all three TORC isoforms was demonstrated by in vitro protein-protein interaction assays and in cells by coimmunoprecipitation that required the N-terminus of TORC and the leucine zipper of dimerized DLK. Overexpressed DLK induced the phosphorylation of TORC2 and TORC1 on Ser-171 and 167, respectively and on additional residues. Since a kinase-dead DLK mutant did not prevent the nuclear localization of TORC and did not reduce TORC transcriptional activity to the same extent as wild-type DLK, we suggest that DLK-induced phosphorylation of TORC contributes to DLK's inhibitory action. Both the interaction with and the phosphorylation of TORC by DLK might account for the reduced recruitment of TORC to a CRE containing promoter as revealed by chromatin immunoprecipitation assay. These results show for the first time the inhibition of TORC function by a mitogen-activated kinase. Given the dependence on TORC in CREB-directed gene transcription, DLK and its downstream kinases thus contribute to the finely tuned regulation of CREB-dependent effects.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , MAP Quinase Quinase Quinases/fisiologia , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Cricetinae , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Fosforilação , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Fatores de Transcrição/genética , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa