Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Pharmacol ; 60(6): 1173-80, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11723223

RESUMO

The physiological implications of lysophosphatidic acid occupancy of individual receptors are largely unknown because selective agonists/antagonists are unavailable currently. The molecular cloning of three high-affinity lysophosphatidic acid receptors, LPA1, LPA2, and LPA3, provides a platform for developing receptor type-selective ligands. Starting with an N-acyl ethanolamide phosphate LPA analog, we made a series of substitutions at the second carbon to generate compounds with varying spatial, stereochemical, and electronic characteristics. Analysis of this series at each recombinant LPA receptor using a guanosine 5'-O-(3-[35S]thio)triphosphate (GTP[gamma35S]) binding assay revealed sharp differences in activity. Our results suggest that these receptors have one spatially restrictive binding pocket that interacts with the 2-substituted moieties and prefers small hydrophobic groups and hydrogen bonding functionalities. The agonist activity predicted by the GTP[gamma35S] binding assay was reflected in the activity of a subset of compounds in increasing arterial pressure in anesthetized rats. One compound with a bulky hydrophobic group (VPC12249) was a dual LPA1/LPA3 competitive antagonist. Several compounds that had smaller side chains were found to be LPA1-selective agonists.


Assuntos
Sistema Cardiovascular/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Receptores de Superfície Celular/antagonistas & inibidores , Receptores Acoplados a Proteínas G , Anestesia , Animais , Pressão Sanguínea/efeitos dos fármacos , Sistema Cardiovascular/fisiopatologia , Células Cultivadas , Humanos , Lisofosfolipídeos/química , Masculino , Conformação Molecular , Ratos , Ratos Wistar , Receptores de Superfície Celular/metabolismo , Receptores de Ácidos Lisofosfatídicos , Relação Estrutura-Atividade
2.
Hypertension ; 38(4): 858-63, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11641299

RESUMO

Obese hypertensives have increased nonesterified fatty acids (NEFAs) and alpha-adrenergic vascular reactivity. Raising NEFAs locally with intralipid and heparin augments dorsal hand venoconstrictor responses to phenylephrine, an alpha(1)-adrenoceptor agonist. The enhanced venoconstrictor responses were reversed by indomethacin. The findings suggest that raising NEFAs leads to the generation of cyclooxygenase (COX) product(s) that enhance vascular reactivity. To test this notion, 6-keto-PGF(1alpha) and TxB(2), the stable metabolites of prostaglandin H(2) (PGH(2)); prostacyclin (PGI(2)); and thromboxane (TxA(2)), were measured approximately 1.5 to 2 cm downstream of a dorsal hand vein infusion of intralipid and heparin (n=10) or saline and heparin (n=5) for 2 hours each. During the third hour, intralipid and heparin (experimental) and saline and heparin (control) were continued, and either saline (control) or indomethacin (intervention) were infused. Intralipid and heparin raised local 6-keto PGF(1alpha) concentrations by 350% to 500% (P<0.005), but saline and heparin did not (P=NS). TxB(2) levels did not change significantly with any infusion. Infusion of indomethacin during the third hour of intralipid and heparin lowered plasma 6-keto-PGF(1alpha) (P<0.05), whereas infusion of saline with intralipid and heparin did not (P=NS). Oleic and linoleic acids at 100 micromol/L, increased 6-keto-PGF(1alpha) in vascular smooth muscle cells (VSMCs) through a protein kinase C and extracellular, signal-regulated kinase independent pathway. However, oleic and linoleic acids increased intracellular Ca(2+) in VSMCs. The data indicate that NEFAs induce the production of COX products, perhaps via Ca(2+)-dependent activation of phospholipase A(2). The COX product(s) may contribute to increased vascular alpha-adrenergic reactivity among insulin-resistant individuals when NEFAs are elevated.


Assuntos
6-Cetoprostaglandina F1 alfa/sangue , Emulsões Gordurosas Intravenosas/farmacologia , Mãos/irrigação sanguínea , Veias/efeitos dos fármacos , 6-Cetoprostaglandina F1 alfa/metabolismo , Adulto , Animais , Cálcio/metabolismo , Células Cultivadas , Feminino , Heparina/farmacologia , Humanos , Indometacina/farmacologia , Ácido Linoleico/farmacologia , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Ácido Oleico/farmacologia , Ácidos Oleicos , Ratos , Ácidos Esteáricos/farmacologia , Tromboxano B2/sangue , Fatores de Tempo
3.
Am J Physiol Heart Circ Physiol ; 281(4): H1545-52, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11557542

RESUMO

Adenylyl cyclases present a potential focal point for signal integration in vascular smooth muscle cells (VSMC) influencing contractile state and cellular responses to vessel wall injury. In the present study, we examined the influence of the vasoactive peptide arginine vasopressin (AVP) on cAMP regulation in primary cultures of rat aortic VSMC and in the A7r5 arterial smooth muscle cell line. In cultured VSMC and A7r5 cells, AVP had no effect on basal cAMP but differentially affected beta-adrenergic receptor-induced activation of adenylyl cyclase. AVP synergistically increased (twofold) isoproterenol-stimulated cAMP production in VSMC but inhibited the effect of isoproterenol (50%) in the A7r5 cell line. The effects of AVP in both preparations were blocked when cells were pretreated with a selective V(1) vasopressin receptor antagonist. Moreover, the actions of AVP in both models were dependent on release of intracellular Ca(2+) and were mimicked by elevation of Ca(2+) with the ionophore A23187, suggesting that the responses to AVP involve Ca(2+)-mediated regulation of adenylyl cyclase stimulation. Adenylyl cyclase types I, III, and VIII are stimulated by Ca(2+)/calmodulin, whereas types V and VI are directly inhibited by Ca(2+). RNA blot analysis for effector isotypes indicated that both VSMC and A7r5 cells expressed types III, V, and VI. VSMC also expressed mRNA for type IV and VIII effectors, which could account for the cell-specific responses to peptide hormone and Ca(2+).


Assuntos
Adenilil Ciclases/metabolismo , Isoenzimas/metabolismo , Músculo Liso Vascular/fisiologia , Transdução de Sinais/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Arginina Vasopressina/farmacologia , Sinalização do Cálcio/fisiologia , Células Cultivadas , AMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Isoproterenol/farmacologia , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/enzimologia , Ratos , Ratos Sprague-Dawley
4.
J Biol Chem ; 276(20): 17339-46, 2001 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-11278760

RESUMO

We used a cultured murine cell model of the inner medullary collecting duct (mIMCD-3 cells) to examine the regulation of the ubiquitous sodium-proton exchanger, Na+/H+ exchanger isoform 1 (NHE-1), by a prototypical G protein-coupled receptor, the bradykinin B2 receptor. Bradykinin rapidly activates NHE-1 in a concentration-dependent manner as assessed by proton microphysiometry of quiescent cells and by 2'-7'-bis[2-carboxymethyl]-5(6)-carboxyfluorescein fluorescence measuring the accelerated rate of pH(i) recovery from an imposed acid load. The activation of NHE-1 is blocked by inhibitors of the bradykinin B2 receptor, phospholipase C, Ca2+/calmodulin (CaM), and Janus kinase 2 (Jak2), but not by pertussis toxin or by inhibitors of protein kinase C and phosphatidylinositol 3'-kinase. Immunoprecipitation studies showed that bradykinin stimulates the assembly of a signal transduction complex that includes CaM, Jak2, and NHE-1. CaM appears to be a direct substrate for phosphorylation by Jak2 as measured by an in vitro kinase assay. We propose that Jak2 is a new indirect regulator of NHE-1 activity, which modulates the activity of NHE-1 by increasing the tyrosine phosphorylation of CaM and most likely by increasing the binding of CaM to NHE-1.


Assuntos
Cálcio/metabolismo , Calmodulina/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas , Receptores da Bradicinina/fisiologia , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Bradicinina/farmacologia , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Concentração de Íons de Hidrogênio , Janus Quinase 2 , Medula Renal/citologia , Medula Renal/fisiologia , Cinética , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Toxina Pertussis , Receptor B2 da Bradicinina , Receptores da Bradicinina/genética , Transdução de Sinais/fisiologia , Vírus 40 dos Símios/genética , Sódio/metabolismo , Transcrição Gênica , Fosfolipases Tipo C/metabolismo , Fatores de Virulência de Bordetella/farmacologia
5.
Pharmacol Ther ; 92(2-3): 179-212, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11916537

RESUMO

The serotonin (5-hydroxytryptamine, 5-HT) receptors have been divided into 7 subfamilies by convention, 6 of which include 13 different genes for G-protein-coupled receptors. Those subfamilies have been characterized by overlapping pharmacological properties, amino acid sequences, gene organization, and second messenger coupling pathways. Post-genomic modifications, such as alternative mRNA splicing or mRNA editing, creates at least 20 more G-protein-coupled 5-HT receptors, such that there are at least 30 distinct 5-HT receptors that signal through G-proteins. This review will focus on what is known about the signaling linkages of the G-protein-linked 5-HT receptors, and will highlight some fascinating new insights into 5-HT receptor signaling.


Assuntos
Receptores de Serotonina/fisiologia , Transdução de Sinais/fisiologia , Adenilil Ciclases/biossíntese , Adenilil Ciclases/farmacologia , AMP Cíclico/metabolismo , Humanos , Canais Iônicos/fisiologia , Proteínas Quinases/biossíntese , Proteínas Quinases/farmacologia , Fosfolipases Tipo C/biossíntese , Fosfolipases Tipo C/farmacologia
6.
Am J Physiol Renal Physiol ; 279(3): F440-8, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10966923

RESUMO

We constructed an expression vector for a fusion protein [ANG II type 1a receptor-green fluorescent protein (AT(1a)R-GFP)] consisting of enhanced GFP attached to the COOH terminus of the rat AT(1a)R. Chinese hamster ovary (CHO) cells transfected with AT(1a)R-GFP demonstrated specific, high-affinity (125)I-labeled ANG II binding (IC(50) 21 nM). ANG II exposure stimulated sodium-proton exchange and cytoplasmic calcium release to a similar extent in cells transfected with AT(1a)R or AT(1a)R-GFP; these responses were desensitized by prior exposure to ANG II and were sensitive to the AT(1)R blocker losartan. ANG II-driven internalization of AT(1a)R-GFP in transfected CHO cells was demonstrated both by radioligand binding and by laser scanning confocal microscopy. Colocalization of GFP fluorescence with that of the nuclear stain TOTO-3 in confocal images was increased more than twofold after 1 h of ANG II exposure. We conclude that AT(1a)R-GFP exhibits similar pharmacological behavior to that of the native AT(1a)R. Our observations also support previous evidence for the presence of AT(1a)R in the nucleus and suggest that the density of AT(1a)R in the nucleus may be regulated by exposure to its ligand.


Assuntos
Núcleo Celular/metabolismo , Indicadores e Reagentes/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Angiotensina II/farmacologia , Animais , Anti-Hipertensivos/farmacologia , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Células CHO , Cálcio/metabolismo , Cricetinae , Corantes Fluorescentes , Genes Reporter , Proteínas de Fluorescência Verde , Radioisótopos do Iodo , Losartan/farmacologia , Microscopia Confocal , Compostos de Quinolínio , Ensaio Radioligante , Ratos , Receptores de Angiotensina/agonistas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Tiazóis
7.
Am J Physiol Gastrointest Liver Physiol ; 278(6): G981-91, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10859229

RESUMO

Clinical studies and in vitro data from isolated parietal cells suggest that acute Helicobacter pylori infection inhibits acid secretion. Gastric acidification is mediated by H(+)-K(+)-ATPase, an integral protein of parietal cell apical membranes. To test the hypothesis that H. pylori downregulates H(+)-K(+)-ATPase alpha-subunit (HKalpha) gene expression and to identify potential intracellular signaling pathways mediating such regulation, we transfected human gastric adenocarcinoma (AGS) cells with human and rat HKalpha 5'-flanking DNA fused to a luciferase reporter plasmid. Histamine caused dose-dependent, cimetidine-sensitive (10(-4) M) increases in cAMP, free intracellular Ca(2+), and HKalpha promoter activation in AGS cells. H. pylori infection of transfected AGS cells dose dependently inhibited basal and histamine-stimulated HKalpha promoter activity by 80% and 66%, respectively. H. pylori dose dependently inhibited phorbol myristate acetate-induced (10(-7) M) and staurosporine- (10(-7) M) and calphostin C-sensitive (5 x 10(-8) M) activation of HKalpha promoter. Also, H. pylori inhibited epidermal growth factor (EGF) (10(-8) M), genistein-sensitive (5 x 10(-5) M) activation of HKalpha promoter, reducing activity to 60% of basal level. These data suggest that H. pylori inhibits HKalpha gene expression via intracellular pathways involving protein kinases A and C and protein tyrosine kinase, AGS cells have functional histamine H(2) and EGF receptors, and transiently transfected AGS cells are a useful model for studying regulation of HKalpha gene expression.


Assuntos
Expressão Gênica/fisiologia , ATPase Trocadora de Hidrogênio-Potássio/genética , Helicobacter pylori/fisiologia , Isoenzimas/genética , Estômago/enzimologia , Animais , Cálcio/metabolismo , Cimetidina/farmacologia , AMP Cíclico/metabolismo , Regulação para Baixo , Infecções por Helicobacter/genética , Histamina/farmacologia , Humanos , Membranas Intracelulares/metabolismo , Regiões Promotoras Genéticas/fisiologia , Ratos , Transfecção , Células Tumorais Cultivadas
8.
Anesth Analg ; 90(5): 1080-5, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10781456

RESUMO

Transient left ventricular (LV) dysfunction can occur after cardioplegic arrest. The contributory mechanisms for this phenomenon are not completely understood. We tested the hypothesis that exposure of LV myocytes to endothelin (ET) during simulated cardioplegic arrest would have direct effects on contractile processes with subsequent reperfusion. LV porcine myocytes were randomly assigned to three groups: 1) CONTROL: normothermic (37 degrees C) cell media (n = 204); 2) Cardioplegia: simulated cardioplegic arrest (K(+) 24 mEq/L, 4 degrees C x 2 h) followed by reperfusion and rewarming with cell media (n = 164); and 3) Cardioplegia/ ET: simulated cardioplegic arrest in the presence of ET (200 pM) followed by reperfusion with cell media containing ET (n = 171). Myocyte contractility was measured by computer-assisted video microscopy. In a subset of experiments, myocyte intracellular calcium was determined after Fluo-3 (Molecular Probes, Eugene, OR) loading by digital fluorescence image analysis. Myocyte shortening velocity was reduced after cardioplegic arrest compared with controls (52 +/- 2 vs 84 +/- 3 microm/s, respectively; P < 0.05) and was further reduced with cardioplegic arrest and ET exposure (43 +/- 2 microm/s, P < 0.05). Intracellular calcium was significantly increased in myocytes exposed to cardioplegia compared with normothermic control myocytes and was further augmented by cardioplegia with ET supplementation (P < 0.05). Exposure of the LV myocyte to ET during cardioplegic arrest directly contributed to contractile dysfunction after reperfusion. Moreover, alterations in intracellular calcium may play a role in potentiating the myocyte contractile dysfunction associated with ET exposure during cardioplegic arrest.


Assuntos
Endotelina-1/farmacologia , Parada Cardíaca Induzida , Contração Miocárdica , Reperfusão Miocárdica , Miocárdio/citologia , Agonistas Adrenérgicos beta/farmacologia , Animais , Cálcio/análise , Ventrículos do Coração/citologia , Técnicas In Vitro , Isoproterenol/farmacologia , Microscopia de Vídeo , Contração Miocárdica/efeitos dos fármacos , Miocárdio/química , Suínos
9.
Biochem J ; 347 Pt 1: 61-7, 2000 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10727402

RESUMO

The hypothesis of this work is that the 'serotonin' or 5-hydroxytryptamine (5-HT)(1A) receptor, which activates the extracellular signal-regulated kinase (ERK) through a G(i)betagamma-mediated pathway, does so through the intermediate actions of reactive oxygen species (ROS). Five criteria were shown to support a key role for ROS in the activation of ERK by the 5-HT(1A) receptor. (1) Antioxidants inhibit activation of ERK by 5-HT. (2) Application of cysteine-reactive oxidant molecules activates ERK. (3) The 5-HT(1A) receptor alters cellular redox properties, and generates both superoxide and hydrogen peroxide. (4) A specific ROS-producing enzyme [NAD(P)H oxidase] is involved in the activation of ERK. (5) There is specificity both in the effects of various chemical oxidizers, and in the putative location of the ROS in the ERK activation pathway. We propose that NAD(P)H oxidase is located in the ERK activation pathway stimulated by the transfected 5-HT(1A) receptor in Chinese hamster ovary (CHO) cells downstream of G(i)betagamma subunits and upstream of or at the level of the non-receptor tyrosine kinase, Src. Moreover, these experiments provide confirmation that the transfected human 5-HT(1A) receptor induces the production of ROS (superoxide and hydrogen peroxide) in CHO cells, and support the possibility that an NAD(P)H oxidase-like enzyme might be involved in the 5-HT-mediated generation of both superoxide and hydrogen peroxide.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NADH NADPH Oxirredutases/metabolismo , Espécies Reativas de Oxigênio/fisiologia , Receptores de Serotonina/fisiologia , Acetilcisteína/farmacologia , Animais , Células CHO , Cricetinae , Fibroblastos/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Cinética , NADPH Oxidases , Oxirredução , Fosforilação , Receptores de Serotonina/genética , Receptores 5-HT1 de Serotonina , Proteínas Recombinantes/metabolismo , Serotonina/farmacologia , Superóxidos/metabolismo , Transfecção , Fatores de Virulência de Bordetella/farmacologia , Domínios de Homologia de src
10.
J Biol Chem ; 275(12): 9026-34, 2000 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-10722752

RESUMO

Gbetagamma binds directly to the third intracellular (i3) loop subdomain of the M(3)-muscarinic receptor (MR). In this report, we identified the Gbetagamma binding motif and G-protein-coupled receptor kinase (GRK2) phosphorylation sites in the M(3)-MR i3 loop via a strategy of deletional and site-directed mutagenesis. The Gbetagamma binding domain was localized to Cys(289)-His(330) within the M(3)-MR-Arg(252)-Gln(490) i3 loop, and the binding properties (affinity, influence of ionic strength) of the M(3)-MR-Cys(289)-His(330) i3 loop subdomain were similar to those observed for the entire i3 loop. Site-directed mutagenesis of the M(3)-MR-Cys(289)-His(330) i3 loop subdomain indicated that Phe(312), Phe(314), and a negatively charged region (Glu(324)-Asp(329)) were required for interaction with Gbetagamma. Generation of the full-length M(3)-MR-Arg(252)-Gln(490) i3 peptides containing the F312A mutation were also deficient in Gbetagamma binding and exhibited a reduced capacity for phosphorylation by GRK2. A similar, parallel strategy resulted in identification of major residues ((331)SSS(333) and (348)SASS(351)) phosphorylated by GRK2, which were just downstream of the Gbetagamma binding motif. Full-length M(3)-MR constructs lacking the 42-amino acid Gbetagamma binding domain (Cys(289)-His(330)) or containing the F312A mutation exhibited ligand recognition properties similar to wild type receptor and also effectively mediated agonist-induced increases in intracellular calcium following receptor expression in Chinese hamster ovary and/or COS 7 cells. However, the M(3)-MRDeltaCys(289)-His(330) and M(3)-MR(F312A) constructs were deficient in agonist-induced sequestration, indicating a key role for the Gbetagamma-M(3)-MR i3 loop interaction in receptor regulation and signal processing.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Subunidades beta da Proteína de Ligação ao GTP , Subunidades gama da Proteína de Ligação ao GTP , Proteínas de Ligação ao GTP/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP , Receptores Muscarínicos/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Transporte Biológico , Proteínas de Ligação ao GTP/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/metabolismo , Fosforilação , Receptor Muscarínico M3 , Receptores Muscarínicos/genética , Proteínas Recombinantes/metabolismo , Serina/metabolismo , Transdução de Sinais , Quinases de Receptores Adrenérgicos beta
11.
Br J Pharmacol ; 127(8): 1751-64, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10482904

RESUMO

The 5-hydroxytryptamine 5-HT1A receptor was one of the first G protein coupled receptors whose cDNA and gene were isolated by molecular cloning methods. Transfection of the cDNA of this receptor into cells previously bearing no 5-HT receptors has resulted in the acquisition of large amounts of information regarding potential signal transduction pathways linked to the receptor, correlations of receptor structure to its various functions, and pharmacological properties of the receptor. Transfection studies with the 5-HT1A receptor have generated critical new information that might otherwise have been elusive. This information notably includes the discovery of unsuspected novel signalling linkages, the elucidation of the mechanisms of receptor desensitization, the refinement of models of the receptor pharmacophore, and the development of silent receptor antagonists, among others. The current review summarizes the most important studies of the recombinant 5-HT1A receptor in the decade since the identification of its cDNA.


Assuntos
Proteínas de Ligação ao GTP/fisiologia , Receptores de Serotonina/fisiologia , Transdução de Sinais/fisiologia , Transfecção/fisiologia , Alelos , Animais , Células HeLa , Humanos , Receptores de Serotonina/química , Receptores de Serotonina/genética , Receptores 5-HT1 de Serotonina , Sistemas do Segundo Mensageiro/fisiologia
12.
Am J Physiol ; 276(6): F922-30, 1999 06.
Artigo em Inglês | MEDLINE | ID: mdl-10362781

RESUMO

We examined the links between fibrotic and proliferative pathways for the 5-HT2A receptor in rat mesangial cells. Serotonin (5-hydroxytryptamine, 5-HT) induced transforming growth factor-beta1 (TGF-beta1) mRNA in a concentration-dependent (peak at 30 nM 5-HT) and time-dependent fashion. For 10 nM 5-HT, the effect was noticeable at 1 h and maximal by 6 h. Inhibition of 1) protein kinase C (PKC), 2) mitogen- and extracellular signal-regulated kinase kinase (MEK1) with 2'-amino-3'-methoxyflavone (PD-90859), and 3) extracellular signal-regulated kinase (ERK) with apigenin attenuated this effect. The effect was blocked by antioxidants, N-acetyl-L-cysteine (NAC) and alpha-lipoic acid, and mimicked by direct application of H2O2. TGF-beta1 mRNA induction was also blocked by diphenyleneiodonium and 4-(2-aminoethyl)-benzenesulfonyl fluoride, which inhibit NAD(P)H oxidase, a source of oxidants. 5-HT increased the amount of TGF-beta1 protein, validating the mRNA studies and demonstrating that 5-HT potently activates ERK and induces TGF-beta1 mRNA and protein in mesangial cells. Mapping studies strongly supported relative positions of the components of the signaling cascade as follow: 5-HT2A receptor --> PKC --> NAD(P)H oxidase/reactive oxygen species --> MEK --> ERK --> TGF-beta1 mRNA. These studies demonstrate that mitogenic signaling components (PKC, MEK, and oxidants) are directly linked to the regulation of TGF-beta1, a key mediator of fibrosis. Thus a single stimulus can direct both proliferative and fibrotic signals in renal mesangial cells.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Mesângio Glomerular/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno , Receptores de Serotonina/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Divisão Celular/fisiologia , Fibrose , Proteínas de Ligação ao GTP/efeitos dos fármacos , Proteínas de Ligação ao GTP/fisiologia , Mesângio Glomerular/citologia , MAP Quinase Quinase 1 , Masculino , NADPH Oxidases/metabolismo , Proteína Quinase C/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Tirosina Quinases/fisiologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/fisiologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/genética , Fatores de Virulência de Bordetella/farmacologia
13.
J Biol Chem ; 274(8): 4749-53, 1999 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-9988712

RESUMO

Many receptors that couple to heterotrimeric guanine nucleotide-binding (G) proteins mediate rapid activation of the mitogen-activated protein kinases, Erk1 and Erk2. The Gi-coupled serotonin (5-hydroxytryptamine (5-HT)) 5-HT1A receptor, heterologously expressed in Chinese hamster ovary or human embryonic kidney 293 cells, mediated rapid activation of Erk1/2 via a mechanism dependent upon both Ras activation and clathrin-mediated endocytosis. This activation was attenuated by chelation of intracellular Ca2+ and Ca2+/calmodulin (CAM) inhibitors or the CAM sequestrant protein calspermin. The CAM-dependent step in the Erk1/2 activation cascade is downstream of Ras activation, because inhibitors of CAM antagonize Erk1/2 activation induced by constitutively activated mutants of Ras and c-Src but not by constitutively activated mutants of Raf and MEK (mitogen and extracellular signal-regulated kinase). Inhibitors of the classical CAM effectors myosin light chain kinase, CAM-dependent protein kinases II and IV, PP2B, and CAM-sensitive phosphodiesterase had no effect upon 5-HT1A receptor-mediated Erk1/2 activation. Because clathrin-mediated endocytosis was required for 5-HT1A receptor-mediated Erk1/2 activation, we postulated a role for CAM in receptor endocytosis. Inhibition of receptor endocytosis by use of sequestration-defective mutants of beta-arrestin1 and dynamin attenuated 5-HT1A receptor-stimulated Erk1/2 activation. Inhibition of CAM prevented agonist-dependent endocytosis of epitope-tagged 5-HT1A receptors. We conclude that CAM-dependent activation of Erk1/2 through the 5-HT1A receptor reflects its role in endocytosis of the receptor, which is a required step in the activation of MEK and subsequently Erk1/2.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Cálcio/metabolismo , Calmodulina/metabolismo , Endocitose , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno , Receptores de Serotonina/metabolismo , Animais , Células CHO , Cálcio/antagonistas & inibidores , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Calmodulina/antagonistas & inibidores , Linhagem Celular , Clatrina/metabolismo , Cricetinae , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Humanos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno , Receptores 5-HT1 de Serotonina , Proteínas Recombinantes/metabolismo
14.
Biochem J ; 330 ( Pt 1): 489-95, 1998 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-9461547

RESUMO

These experiments tested the hypothesis that signalling elements involved in the activation of the extracellular signal-regulated protein kinase (ERK) mediate rapid activation of sodium-proton exchange (NHE) in fibroblasts when both signals are initiated by a single G protein-coupled receptor, the 5-HT1A receptor. Similarities between the two processes were comparable concentration-response curves and time-courses, and overlapping sensitivity to some pharmacological inhibitors of tyrosine kinases (staurosporine and genistein), and phosphoinositide 3'-kinase (wortmannin and LY204002). Activation of NHE was much more sensitive to the phosphatidylcholine-specific phospholipase inhibitor (D609) than was ERK. Neither pathway was sensitive to manoeuvres designed to block PKC. In contrast, Src or related kinases appear to be required to activate ERK, but not NHE. Transfection of cDNA constructs encoding inactive mutant phosphoinositide 3'-kinase, Grb2, Sos, Ras, and Raf molecules were successful in attenuating ERK, but had essentially no effect upon NHE activation. Finally, PD98059, an inhibitor of mitogen activated/extracellular signal regulated kinase kinase, blocked ERK but not NHE activation. Thus, in CHO fibroblast cells, activation by the 5-HT1A receptor of ERK and NHE share a number of overlapping features. However, our studies do not support a major role for ERK, when activated by the 5-HT1A receptor, as a short-term upstream regulator of NHE activity.


Assuntos
Proteínas de Ligação ao GTP/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Receptores de Serotonina/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Células CHO , Cricetinae , Fibroblastos , Humanos , Fosfolipídeos/fisiologia , Proteína Quinase C/fisiologia , Proteínas Tirosina Quinases/metabolismo , Receptores 5-HT1 de Serotonina , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...