Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 229
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Br J Pharmacol ; 150(3): 255-7, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17200675

RESUMEN

Sphingosine kinase-1 (SphK1) catalyses the phosphorylation of sphingosine to sphingosine-1-phosphate (S1P), which acts on at least five specific G-protein-coupled receptors and also intracellularly. SphK1 has been implicated in cell proliferation, cancer growth, chemoresistance, immune cell functions and cell migration. In this issue of the British Journal of Pharmacology, Klawitter et al. demonstrate that extracellular nucleotides stimulate the migration of renal mesangial cells. The nucleotides furthermore upregulated SphK1 expression and activity, and this enzyme was required for nucleotide-induced migration. Together with previous findings, these data raise exciting questions: by which mechanism does SphK1 regulate migration in mesangial cells, how is the interplay of purinoceptors and S1P receptors organized in these cells, and how would SphK1-deficient mice respond to kidney damage?


Asunto(s)
Movimiento Celular/fisiología , Células Mesangiales/enzimología , Nucleótidos/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Ratones , Receptores de Lisoesfingolípidos , Receptores Purinérgicos
3.
FEBS Lett ; 509(2): 239-44, 2001 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-11741596

RESUMEN

Formation of sphingosine-1-phosphate (SPP) by sphingosine kinase serves as a signalling pathway for various membrane receptors. Here, we show that membrane depolarisation is another mechanism by which this pathway can be activated. Formation of [(3)H]SPP as well as levels of endogenous SPP were rapidly and transiently increased in PC12 pheochromocytoma cells depolarised with high KCl. Time course and maximum were similar to those induced by bradykinin. Depolarisation-induced SPP production was also observed in RINm5F insulinoma cells, dependent on extracellular Ca(2+) and fully suppressed by verapamil, thus apparently caused by Ca(2+) influx via voltage-gated Ca(2+) channels. Studies with sphingosine kinase inhibitors and overexpression of sphingosine kinase revealed a partial contribution of this pathway to depolarisation-induced noradrenaline release and Ca(2+) increase.


Asunto(s)
Canales de Calcio/metabolismo , Membrana Celular/metabolismo , Lisofosfolípidos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Animales , Bradiquinina/farmacología , Señalización del Calcio , Norepinefrina/metabolismo , Células PC12 , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Cloruro de Potasio/farmacología , Ratas , Proteínas Recombinantes/metabolismo , Esfingosina/farmacología , Verapamilo/farmacología
4.
Nat Cell Biol ; 3(11): 1020-4, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11715024

RESUMEN

Stimulation of phosphoinositide-hydrolysing phospholipase C (PLC) generating inositol-1,4,5-trisphosphate is a major calcium signalling pathway used by a wide variety of membrane receptors, activating distinct PLC-beta or PLC-gamma isoforms. Here we report a new PLC and calcium signalling pathway that is triggered by cyclic AMP (cAMP) and mediated by a small GTPase of the Rap family. Activation of the adenylyl cyclase-coupled beta2-adrenoceptor expressed in HEK-293 cells or the endogenous receptor for prostaglandin E1 in N1E-115 neuroblastoma cells induced calcium mobilization and PLC stimulation, seemingly caused by cAMP formation, but was independent of protein kinase A (PKA). We provide evidence that these receptor responses are mediated by a Rap GTPase, specifically Rap2B, activated by a guanine-nucleotide-exchange factor (Epac) regulated by cAMP, and involve the recently identified PLC-epsilon isoform.


Asunto(s)
Señalización del Calcio , AMP Cíclico/metabolismo , Fosfolipasas de Tipo C/metabolismo , Proteínas de Unión al GTP rap/metabolismo , Adenilil Ciclasas/metabolismo , Alprostadil/metabolismo , Línea Celular , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Fosfoinositido Fosfolipasa C , Receptores Adrenérgicos beta/genética , Receptores Adrenérgicos beta/metabolismo , Células Tumorales Cultivadas , Fosfolipasas de Tipo C/genética
5.
Life Sci ; 68(22-23): 2535-40, 2001 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-11392623

RESUMEN

Based on the finding that G protein-coupled receptors (GPCRs) can induce Ca2+ mobilization, apparently independent of the phospholipase C (PLC)/inositol-1,4,5-trisphosphate (IP3) pathway, we investigated whether sphingosine kinase, which generates sphingosine-1-phosphate (SPP), is involved in calcium signaling by mAChR and other GPCRs. Inhibition of sphingosine kinase by DL-threo-dihydrosphingosine and N,/N-dimethylsphingosine markedly inhibited [Ca2+]i increases elicited by M2 and M3 mAChRs in HEK-293 cells without affecting PLC activation. Activation of M2 and M3 mAChR rapidly and transiently stimulated production of SPP. Furthermore, microinjection of SPP into HEK-293 cells induced rapid and transient Ca2+ mobilization. Pretreatment of HEK-293 cells with the calcium chelator BAPTA/AM fully blocked mAChR-induced SPP production. On the other hand, incubation of HEK-293 cells with calcium ionophores activated SPP production. Similar findings were obtained for formyl peptide and P2Y2 purinergic receptors in HL-60 cells. On the basis of these studies we propose, that following initial IP3 production by receptor-mediated PLC activation, a local discrete increase in [Ca2+]i induces sphingosine kinase stimulation, which ultimately leads to full calcium mobilization. Thus, sphingosine kinase activation most likely represents an amplification system for calcium signaling by mAChRs and other GPCRs.


Asunto(s)
Señalización del Calcio , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores Muscarínicos/metabolismo , Esfingosina/metabolismo , Carbacol/farmacología , Línea Celular , Agonistas Colinérgicos/farmacología , Inhibidores Enzimáticos/metabolismo , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Ensayo de Unión Radioligante , Esfingosina/análogos & derivados
6.
Br J Pharmacol ; 132(8): 1925-33, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11309265

RESUMEN

Lysosphingolipids such as sphingosine-1-phosphate (SPP) and sphingosylphosphorylcholine (SPPC) can act on specific G-protein-coupled receptors. Since SPP and SPPC cause renal vasoconstriction, we have investigated their effects on urine and electrolyte excretion in anaesthetized rats. Infusion of SPP (1 - 30 microg kg(-1) min(-1)) for up to 120 min dose-dependently but transiently (peak after 15 min, disappearance after 60 min) reduced renal blood flow without altering endogenous creatinine clearance. Nevertheless, infusion of SPP increased diuresis, natriuresis and calciuresis and, to a lesser extent, kaliuresis. These tubular lysosphingolipid effects developed more slowly (maximum after 60 - 90 min) and also abated more slowly upon lysosphingolipid washout than the renovascular effects. Infusion of SPPC, sphingosine and glucopsychosine (3 - 30 microg kg(-1) min(-1) each) caused little if any alterations in renal blood flow but also increased diuresis, natriuresis and calciuresis and, to a lesser extent, kaliuresis. Pretreatment with pertussis toxin (10 microg kg(-1) 3 days before the acute experiment) abolished the renovascular and tubular effects of 30 microg kg(-1) min(-1) SPP. These findings suggest that lysosphingolipids are a hitherto unrecognized class of endogenous modulators of renal function. SPP affects renovascular tone and tubular function via receptors coupled to G(i)-type G-proteins. SPPC, sphingosine and glucopsychosine mimic only the tubular effects of SPP, and hence may act on distinct sites.


Asunto(s)
Diuresis/fisiología , Natriuresis/fisiología , Psicosina/análogos & derivados , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G , Anestesia , Animales , Presión Sanguínea/efectos de los fármacos , Diuresis/efectos de los fármacos , Túbulos Renales/efectos de los fármacos , Túbulos Renales/fisiología , Masculino , Natriuresis/efectos de los fármacos , Toxina del Pertussis , Psicosina/metabolismo , Ratas , Ratas Wistar , Receptores de Superficie Celular/efectos de los fármacos , Receptores Lisofosfolípidos , Circulación Renal/efectos de los fármacos , Circulación Renal/fisiología , Esfingolípidos/farmacología , Factores de Virulencia de Bordetella/farmacología
7.
Eur J Pharmacol ; 414(2-3): 145-54, 2001 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-11239914

RESUMEN

Recently, a family of G-protein-coupled receptors named endothelial differentiation gene (Edg) receptor family has been identified, which are specifically activated by the two serum lipids, sphingosine-1-phosphate and lysophosphatidic acid. Sphingosine-1-phosphate can also act intracellularly to release Ca2+ from intracellular stores. Since in several cell types, G-protein-coupled lysophosphatidic acid or sphingosine-1-phosphate receptors mobilize Ca2+ in the absence of a measurable phospholipase C stimulation, it was analysed here whether intracellular sphingosine-1-phosphate production was the signalling mechanism used by extracellular sphingosine-1-phosphate for mobilization of stored Ca2+. Sphingosine-1-phosphate and the low affinity sphingosine-1-phosphate receptor agonist, sphingosylphosphorylcholine, induced a rapid, transient and nearly complete pertussis toxin-sensitive Ca2+ mobilization in human embryonic kidney (HEK-293) cells. The G-protein-coupled sphingosine-1-phosphate receptors, Edg-1, Edg-3 and Edg-5, were found to be endogenously expressed in these cells. Most interestingly, sphingosine-1-phosphate and sphingosylphosphorylcholine did not induce a measurable production of inositol-1,4,5-trisphosphate or accumulation of inositol phosphates. Instead, sphingosine-1-phosphate and sphingosylphosphorylcholine induced a rapid and transient increase in production of intracellular sphingosine-1-phosphate with a maximum of about 1.4-fold at 30 s. Stimulation of sphingosine-1-phosphate formation by sphingosine-1-phosphate and sphingosylphosphorylcholine was fully blocked by pertussis toxin, indicating that extracellular sphingosine-1-phosphate via endogenously expressed G(i)-coupled receptors induces a stimulation of intracellular sphingosine-1-phosphate production. As sphingosine-1-phosphate- and sphingosylphosphorylcholine-induced increases in intracellular Ca2+ were blunted by sphingosine kinase inhibitors, this sphingosine-1-phosphate production appears to mediate Ca2+ signalling by extracellular sphingosine-1-phosphate and sphingosylphosphorylcholine in HEK-293 cells.


Asunto(s)
Señalización del Calcio/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas I-kappa B , Proteínas Inmediatas-Precoces/metabolismo , Lisofosfolípidos , Fosforilcolina/análogos & derivados , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Señalización del Calcio/efectos de los fármacos , Carbacol/farmacología , Línea Celular , Agonistas Colinérgicos/farmacología , Proteínas de Unión al GTP/efectos de los fármacos , Proteínas de Unión al GTP/metabolismo , Humanos , Inhibidor NF-kappaB alfa , Fosforilcolina/farmacología , Fosfotransferasas (Aceptor de Grupo Alcohol)/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Receptores de Superficie Celular/efectos de los fármacos , Receptores Lisofosfolípidos , Esfingosina/farmacología , Fosfolipasas de Tipo C/efectos de los fármacos , Fosfolipasas de Tipo C/metabolismo
8.
J Biol Chem ; 276(4): 2474-9, 2001 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-11036069

RESUMEN

The M(3) muscarinic acetylcholine receptor (mAChR) expressed in HEK-293 cells couples to G(q) and G(12) proteins and stimulates phospholipase C (PLC) and phospholipase D (PLD) in a pertussis toxin-insensitive manner. To determine the type of G protein mediating M(3) mAChR-PLD coupling in comparison to M(3) mAChR-PLC coupling, we expressed various Galpha proteins and regulators of the G protein signaling (RGS), which act as GTPase-activating proteins for G(q)- or G(12)-type G proteins. PLD stimulation by the M(3) mAChR was enhanced by the overexpression of Galpha(12) and Galpha(13), whereas the overexpression of Galpha(q) strongly increased PLC activity without affecting PLD activity. Expression of the RGS homology domain of Lsc, which acts specifically on Galpha(12) and Galpha(13), blunted the M(3) mAChR-induced PLD stimulation without affecting PLC stimulation. On the other hand, overexpression of RGS4, which acts on Galpha(q)- but not Galpha(12)-type G proteins, suppressed the M(3) mAChR-induced PLC stimulation without altering PLD stimulation. We conclude that the M(3) mAChR in HEK-293 cells apparently signals to PLD via G(12)- but not G(q)-type G proteins and that G protein subtype-selective RGS proteins can be used as powerful tools to dissect the pertussis toxin-resistant G proteins and their role in receptor-effector coupling.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas/metabolismo , Fosfolipasa D/metabolismo , Receptores Muscarínicos/metabolismo , Carbacol/farmacología , Resistencia a Medicamentos , Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Humanos , Agonistas Muscarínicos/farmacología , Toxina del Pertussis , Proteínas RGS/metabolismo , Receptor Muscarínico M3 , Transducción de Señal , Fosfolipasas de Tipo C/metabolismo , Factores de Virulencia de Bordetella/farmacología
9.
Biochem J ; 352 Pt 2: 319-25, 2000 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11085924

RESUMEN

Rho GTPases are implicated in a multitude of cellular processes regulated by membrane receptors, such as cytoskeletal rearrangements, gene transcription and cell growth and motility. Activation of these GTPases is under the direct control of guanine nucleotide exchange factors (GEFs), the Dbl family proteins. By searching protein databases we have identified a novel Rho-GEF, termed p114-Rho-GEF, which similarly to other Rho-GEFs contains a Dbl homology domain followed by a pleckstrin homology domain. p114-Rho-GEF interacted specifically with RhoA, in its nucleotide-free and guanosine 5'-[gamma-thio]triphosphate-bound states, but not with Rac1 and Cdc42, and efficiently catalysed guanine nucleotide exchange of RhoA. Consistent with these results in vitro was our finding that the overexpression of p114-Rho-GEF in J82 and HEK-293 cells induced the formation of actin stress fibres and stimulated serum-response-factor-mediated gene transcription in a Rho-dependent manner. Rho-mediated transcriptional activation induced by M(3) muscarinic acetylcholine and lysophosphatidic acid receptors was enhanced by p114-Rho-GEF, suggesting that the activity of this novel Rho-GEF, which is widely expressed in human tissues, can be controlled by G-protein-coupled receptors.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , ADN Complementario , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/fisiología , Humanos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Activación Transcripcional/fisiología
10.
Mol Pharmacol ; 58(3): 491-7, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10953041

RESUMEN

Sphingosine-1-phosphate (SPP), produced by sphingosine kinase, has recently been reported to act as an intracellular second messenger for Ca(2+) and mitogenic responses triggered by membrane receptors and as an extracellular ligand for specific SPP receptors. Here, we investigated the signaling pathway leading to SPP production by the G protein-coupled P2Y(2) receptor and its functional implication in human leukemia (HL-60) cells, which do not respond to extracellular SPP. P2Y(2) receptor activation by UTP or ATP resulted in rapid and transient production of SPP, which was insensitive to pertussis toxin and blocked by the sphingosine kinase inhibitor, DL-threo-dihydrosphingosine. Treatment of HL-60 cells with this inhibitor did not affect activation of mitogen-activated protein kinases, but suppressed Ca(2+) mobilization by the P2Y(2) receptor. However, receptor-induced SPP production apparently required an increase in intracellular Ca(2+) concentration, but not Ca(2+) influx, and was mimicked by exposure of cells to Ca(2+) ionophores. Taken together, activation of the P2Y(2) receptor stimulates SPP production in HL-60 cells, a process apparently not required for mitogen-activated protein kinase activation, but most likely representing an amplification system for receptor-mediated Ca(2+) signaling.


Asunto(s)
Calcio/metabolismo , Lisofosfolípidos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Receptores Purinérgicos P2/metabolismo , Esfingosina/análogos & derivados , Esfingosina/biosíntesis , Transporte Biológico , Activación Enzimática , Células HL-60 , Humanos , Receptores Purinérgicos P2Y2
11.
Br J Pharmacol ; 130(8): 1878-83, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10952678

RESUMEN

Sphingolipids such as sphingosine-1-phosphate (SPP) and sphingosylphosphorylcholine constrict isolated rat intrarenal and mesenteric microvessels in vitro. The present study investigates their effects on the cardiovascular system in vivo in anaesthetized rats. The animals were given intravenous or intrarenal arterial bolus injections of sphingolipids (0.1-100 microg kg(-1)) with subsequent measurements of mean arterial pressure, heart rate and renal and mesenteric blood flows (RBF, MBF) using a pressure transducer and electromagnetic flow probes, respectively. Intravenous injection of SPP rapidly (within 30 s), transiently and dose-dependently reduced RBF (maximally -4.0+/-0.3 ml min(-1)) and MBF (maximally -1.4+/-0.2 ml min(-1)), without affecting mean arterial pressure or heart rate. Other sphingolipids had no significant effect. Intrarenal arterial SPP administration caused greater blood flow reductions (maximally -6.4+/-0.3 ml min(-1)) than systemic administration. Upon intrarenal administration, sphingosylphos- phorylcholine also lowered RBF (maximally -2.8+/-0.6 ml min(-1)), while the other sphingolipids remained without effect. Pretreatment with pertussis toxin (PTX, 10 microg kg(-1)) 3 days before the acute experiment abolished the SPP-induced reductions of RBF and MBF. These data demonstrate, that SPP is a potent vasoconstrictor in vivo, particularly in the renal vasculature, while the other structurally related sphingolipids had little if any effects. The PTX-sensitivity strongly suggests that the effects of SPP on renal and mesenteric blood flow are mediated by receptors coupled to G(i)-type G-proteins.


Asunto(s)
Lisofosfolípidos , Toxina del Pertussis , Fosforilcolina/análogos & derivados , Psicosina/análogos & derivados , Circulación Renal/efectos de los fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacología , Circulación Esplácnica/efectos de los fármacos , Factores de Virulencia de Bordetella/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Frecuencia Cardíaca/efectos de los fármacos , Riñón/irrigación sanguínea , Masculino , Mesenterio/irrigación sanguínea , Fosforilcolina/farmacología , Psicosina/farmacología , Ratas , Ratas Wistar
12.
Br J Pharmacol ; 130(8): 1871-7, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10952677

RESUMEN

Sphingolipids such as sphingosine-1-phosphate (SPP) and sphingosylphosphorylcholine (SPPC) can act both intracellularly and at G-protein-coupled receptors, some of which were cloned and designated as Edg-receptors. Sphingolipid-induced vascular effects were determined in isolated rat mesenteric and intrarenal microvessels. Additionally, sphingolipid-induced elevations in intracellular Ca(2+) concentration were measured in cultured rat aortic smooth muscle cells. SPPC and SPP (0.1-100 micromol l(-1)) caused concentration-dependent contraction of mesenteric and intrarenal microvessels (e.g. SPPC in mesenteric microvessels pEC(50) 5.63+/-0.17 and E(max) 49+/-3% of noradrenaline), with other sphingolipids being less active. The vasoconstrictor effect of SPPC in mesenteric microvessels was stereospecific (pEC(50) D-erythro-SPPC 5.69+/-0.08, L-threo-SPPC 5.31+/-0.06) and inhibited by pretreatment with pertussis toxin (E(max) from 44+/-5 to 19+/-4%), by chelation of extracellular Ca(2+) with EGTA and by nitrendipine (E(max) from 40+/-6 to 6+/-1 and 29+/-6%, respectively). Mechanical endothelial denudation or NO synthase inhibition did not alter the SPPC effects, while indomethacin reduced them (E(max) from 87+/-3 to 70+/-4%). SPP and SPPC caused transient increases in intracellular Ca(2+) concentrations in rat aortic smooth muscle cells in a pertussis toxin-sensitive manner. Our data demonstrate that SPP and SPPC cause vasoconstriction of isolated rat microvessels and increase intracellular Ca(2+) concentrations in cultured rat aortic smooth muscle cells. These effects appear to occur via receptors coupled to pertussis toxin-sensitive G-proteins. This is the first demonstration of effects of SPP and SPPC on vascular tone and suggests that sphingolipids may be an hitherto unrecognized class of endogenous regulators of vascular tone.


Asunto(s)
Lisofosfolípidos , Arterias Mesentéricas/efectos de los fármacos , Fosforilcolina/análogos & derivados , Fosforilcolina/farmacología , Psicosina/análogos & derivados , Arteria Renal/efectos de los fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacología , Vasoconstricción/efectos de los fármacos , Animales , Calcio/metabolismo , Relación Dosis-Respuesta a Droga , Ácido Egtácico/farmacología , Técnicas In Vitro , Indometacina/farmacología , Masculino , Arterias Mesentéricas/fisiología , Nitrendipino/farmacología , Nitroarginina/farmacología , Toxina del Pertussis , Psicosina/farmacología , Ratas , Ratas Wistar , Arteria Renal/fisiología , Vasodilatadores/farmacología , Factores de Virulencia de Bordetella/farmacología
13.
Eur J Biochem ; 267(16): 5237-46, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10931209

RESUMEN

The involvement of small GTPases of the Rho family in the control of phosphoinositide metabolism by adhesion signals was examined in NIH 3T3 fibroblasts. Abrogation of adhesion signals by detachment of cells from their substratum resulted in a time-dependent decrease in the cellular level of PtdIns(4,5)P2 by approximately 50%. This effect could be mimicked by treatment of adherent cells with Clostridium difficile toxin B and toxin B-1470, which inhibit specific subsets of Rho and Ras GTPases. Detachment of cells that had been pretreated with the clostridial toxins did not cause a further reduction in PtdIns(4,5)P2 levels, suggesting that the target GTPases are integrated into the control of phosphoinositide levels by adhesion signals. The reduction in PtdIns(4,5)P2 levels could be attributed to reduced activity of the major PtdIns(4, 5)P2-producing enzyme, PtdIns4P 5-kinase. Unexpectedly, both cell detachment and toxin treatment resulted in a twofold to threefold increase in inositol phosphate production in intact cells. In lysates of these cells, in vitro phospholipase C activity was found to be elevated by 30-50%. The effects of cell detachment and toxin treatment on inositol phosphate formation could be mimicked by expression of dominant-negative N17 Rac1. Taken together, these data suggest that adhesion-controlled small GTPases of the Rho family are involved in the regulation of the cellular PtdIns(4,5)P2 levels in NIH 3T3 fibroblasts, by controlling the activities of both PtdIns4P 5-kinase and phospholipase C.


Asunto(s)
Proteínas Bacterianas , Adhesión Celular/fisiología , Fosfatidilinositol 4,5-Difosfato/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Fosfolipasas de Tipo C/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Células 3T3 , Animales , Toxinas Bacterianas/farmacología , Calcio/fisiología , Adhesión Celular/efectos de los fármacos , Citotoxinas/farmacología , Cinética , Ratones , Fosfoinositido Fosfolipasa C , Fosforilación , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transfección , Proteína de Unión al GTP rac1/metabolismo
14.
Mol Pharmacol ; 58(2): 449-54, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10908314

RESUMEN

Sphingosine-1-phosphate (SPP) and sphingosylphosphorylcholine (SPPC) have been reported to activate muscarinic receptor-activated inward rectifier K(+) current (I(K.ACh)) in cultured guinea pig atrial myocytes with similar nanomolar potency. Members of the endothelial differentiation gene (Edg) receptor family were recently identified as receptors for SPP; however, these receptors respond only to micromolar concentrations of SPPC. Here we investigated the sphingolipid-induced activation of I(K.ACh) in freshly isolated guinea pig, mouse, and human atrial myocytes. SPP activated I(K.ACh) in atrial myocytes from all three species with a similar nanomolar potency (EC(50) values: 4-8 nM). At these low concentrations, SPPC also activated I(K.ACh) in guinea pig myocytes. In contrast, SPPC was almost ineffective in mouse and human myocytes, thus resembling the pharmacology of the Edg receptors. Transcripts of Edg-1, Edg-3, and Edg-5 were detected in human atrial cells. Moreover, activation of I(K.ACh) by SPP was blocked by the Edg-3-selective antagonist suramin, which did not affect basal or carbachol-stimulated K(+) currents. In conclusion, these data indicate that I(K.ACh) activation by SPP and SPPC exhibits large species differences. Furthermore, they suggest that SPP-induced I(K.ACh) activation in human atrial myocytes is mediated by the Edg-3 subtype of SPP receptors.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas I-kappa B , Lisofosfolípidos , Miocardio/metabolismo , Canales de Potasio de Rectificación Interna , Canales de Potasio/metabolismo , Esfingosina/análogos & derivados , Esfingosina/farmacología , Análisis de Varianza , Animales , Femenino , Cobayas , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/metabolismo , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibidor NF-kappaB alfa , Receptores Lisofosfolípidos , Especificidad de la Especie
15.
J Biol Chem ; 275(42): 32603-10, 2000 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-10908568

RESUMEN

Activation of stably expressed M(2) and M(3) muscarinic acetylcholine receptors (mAChRs) as well as of endogenously expressed lysophosphatidic acid and purinergic receptors in HEK-293 cells can induce a long lasting potentiation of phospholipase C (PLC) stimulation by these and other G protein-coupled receptors (GPCRs). Here, we report that GPCRs can induce an up-regulation of PLC stimulation by receptor tyrosine kinases (RTKs) as well and provide essential mechanistic characteristics of this sensitization process. Pretreatment of HEK-293 cells for 2 min with carbachol, a mAChR agonist, lysophosphatidic acid, or ATP, followed by agonist washout, strongly increased (by 2-3-fold) maximal PLC stimulation (measured >/=40 min later) by epidermal growth factor and platelet-derived growth factor, but not insulin, and largely enhanced PLC sensitivity to these RTK agonists. The up-regulation of RTK-induced PLC stimulation was cycloheximide-insensitive and was observed for up to approximately 90 min after removal of the GPCR agonist. Sensitization of receptor-induced PLC stimulation caused by prior M(2) mAChR activation was fully prevented by pertussis toxin and strongly reduced by expression of Gbetagamma scavengers. Furthermore, inhibition of conventional protein kinase C (PKC) isoenzymes and chelation of intracellular Ca(2+) suppressed the sensitization process, while overexpression of PKC-alpha, but not PKC-betaI, further enhanced the M(2) mAChR-induced sensitization of PLC stimulation. None of these treatments affected acute PLC stimulation by either GPCR or RTK agonists. Taken together, short term activation of GPCRs can induce a strong and long lasting sensitization of PLC stimulation by RTKs, a process apparently involving G(i)-derived Gbetagammas as well as increases in intracellular Ca(2+) and activation of a PKC isoenzyme, most likely PKC-alpha.


Asunto(s)
Carbacol/farmacología , Factor de Crecimiento Epidérmico/farmacología , Proteínas de Unión al GTP/fisiología , Fosfatos de Inositol/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Muscarínicos/fisiología , Fosfolipasas de Tipo C/metabolismo , Adenosina Trifosfato/farmacología , Línea Celular , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Isoenzimas/metabolismo , Cinética , Proteína Quinasa C/metabolismo , Proteína Quinasa C beta , Proteína Quinasa C-alfa , Receptor Muscarínico M2 , Receptor Muscarínico M3 , Proteínas Recombinantes/metabolismo , Transfección
16.
J Biol Chem ; 275(29): 21969-74, 2000 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-10801820

RESUMEN

Most G protein-coupled receptors (GPCRs), including the M(1) muscarinic acetylcholine receptor (mAChR), internalize in clathrin-coated vesicles, a process that requires dynamin GTPase. The observation that some GPCRs like the M(2) mAChR and the angiotensin AT(1A) receptor (AT(1A)R) internalize irrespective of expression of dominant-negative K44A dynamin has led to the proposal that internalization of these GPCRs is dynamin-independent. Here, we report that, contrary to what is postulated, internalization of M(2) mAChR and AT(1A)R in HEK-293 cells is dynamin-dependent. Expression of N272 dynamin, which lacks the GTP-binding domain, or K535M dynamin, which is not stimulatable by phosphatidylinositol 4, 5-bisphosphate, strongly inhibits internalization of M(1) and M(2) mAChRs and AT(1A)Rs. Expression of kinase-defective K298M c-Src or Y231F,Y597F dynamin (which cannot be phosphorylated by c-Src) reduces M(1) mAChR internalization. Similarly, c-Src inhibitor PP1 as well as the generic tyrosine kinase inhibitor genistein strongly inhibit M(1) mAChR internalization. In contrast, M(2) mAChR internalization is not (or is only slightly) reduced by expression of these constructs or treatment with PP1 or genistein. Thus, dynamin GTPases are not only essential for M(1) mAChR but also for M(2) mAChR and AT(1A)R internalization in HEK-293 cells. Our findings also indicate that dynamin GTPases are differentially regulated by c-Src-mediated tyrosine phosphorylation.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Receptores de Angiotensina/metabolismo , Receptores Muscarínicos/metabolismo , Transducción de Señal , Animales , Línea Celular , Dinaminas , Ratas
17.
J Biol Chem ; 275(14): 10168-74, 2000 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-10744700

RESUMEN

The serine/threonine kinase Rho-kinase was recently identified as a downstream effector of the small GTPase Rho, mediating effects of Rho on the actin cytoskeleton. Also phosphatidylinositol 4,5-bisphosphate (PI(4,5)P(2)) has been implicated in the regulation of actin polymerization. As the synthesis of PI(4,5)P(2) has been suggested to be affected by Rho proteins, we investigated whether Rho-kinase is involved in the control of PI(4,5)P(2) levels. Overexpression of RhoA in HEK-293 cells increased phosphatidylinositol 4-phosphate (PI4P) 5-kinase activity and concomitantly enhanced cellular PI(4,5)P(2) levels, whereas overexpression of the Rho-inactivating C3 transferase decreased both PI4P 5-kinase activity and PI(4,5)P(2) levels. These effects of RhoA could be mimicked by overexpression of wild-type Rho-kinase and of the constitutively active catalytic domain of Rho-kinase, Rho-kinase-CAT. In contrast, a kinase-deficient mutant of Rho-kinase had no effect on PI4P 5-kinase activity. Importantly, the increase in PI4P 5-kinase activity and PI(4,5)P(2) levels by wild-type Rho-kinase, but not by Rho-kinase-CAT, was completely prevented by coexpression of C3 transferase, indicating that the effect of Rho-kinase was under the control of endogenous Rho. In cell lysates, addition of recombinant RhoA and Rho-kinase-CAT stimulated PI4P 5-kinase activity. Finally, the increase in PI(4,5)P(2) levels induced by both Rho-kinase-CAT and RhoA was reversed by the Rho-kinase inhibitor HA-1077. Our data suggest that Rho-kinase is involved in the Rho-controlled synthesis of PI(4,5)P(2) by PI4P 5-kinase.


Asunto(s)
Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Línea Celular , Cloranfenicol O-Acetiltransferasa/genética , Activación Enzimática , Humanos , Péptidos y Proteínas de Señalización Intracelular , Cinética , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilinositoles/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Quinasas Asociadas a rho , Proteína de Unión al GTP rhoA/genética
18.
J Neurochem ; 74(4): 1721-30, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10737631

RESUMEN

Although M1-M4 muscarinic acetylcholine receptors (mAChRs) in HEK-293 cells internalize on agonist stimulation, only M1, M3, and M4 but not M2 mAChRs recycle to the plasma membrane. To investigate the functional consequences of this phenomenon, we compared desensitization and resensitization of M2 versus M4 mAChRs. Treatment with 1 mM carbachol for 1 h at 37 degrees C reduced numbers of cell surface M2 and M4 mAChRs by 40-50% and M2 and M4 mAChR-mediated inhibition of adenylyl cyclase, intracellular Ca2+ concentration ([Ca2+]i) increases, and phospholipase C (PLC) activation by 60-70%. Receptor-mediated inhibition of adenylyl cyclase and [Ca2+]i increases significantly resensitized within 3 h. However, M4 but not M2 mAChR-mediated PLC activation resensitized. At 16 degrees C, M2 mAChR-mediated [Ca2+]i increases and PLC stimulation desensitized to a similar extent as at 37 degrees C. However, at 16 degrees C, where M2 mAChR internalization is negligible, both M2 mAChR responses resensitized, demonstrating that M2 mAChR resensitization proceeds at the plasma membrane. Examination of M2 mAChR responses following inactivation of cell surface mAChRs by quinuclidinyl benzilate revealed substantial receptor reserve for coupling to [Ca2+]i increases but not to PLC. We conclude that M2 mAChR internalization induces long-lasting PLC desensitization predominantly because receptor loss is not compensated for by receptor recycling or receptor reserve.


Asunto(s)
Receptores Muscarínicos/metabolismo , Transducción de Señal/fisiología , Fosfolipasas de Tipo C/metabolismo , Calcio/metabolismo , Carbacol/farmacología , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Agonistas Colinérgicos/farmacología , Colforsina/farmacología , AMP Cíclico/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Endocitosis/fisiología , Humanos , Riñón/citología , Cinética , Receptor Muscarínico M2 , Receptor Muscarínico M4 , Receptores de Superficie Celular/metabolismo , Transfección , Tritio
19.
Naunyn Schmiedebergs Arch Pharmacol ; 361(1): 1-11, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10651140

RESUMEN

Sphingosine-1-phosphate (SPP) induces a variety of cellular responses, including Ca2+ signaling, proliferation, and inhibition of motility, apparently by acting at specific G protein coupled receptors. Here, the expression, signaling, and motile responses of sphingolipid receptors were examined in human bladder carcinoma (J82) cells, for which lysophosphatidic acid (LPA) and thrombin act as potent agonists. SPP potently and rapidly mobilized Ca2+, stimulated phospholipases C and D, and inhibited cAMP accumulation, without affecting growth of J82 cells, which express the recently identified SPP receptors, Edg-1 and Edg-3. The effects of SPP were mimicked by sphingosylphosphorylcholine (SPPC) and strongly attenuated by pertussis toxin (PTX). SPP and SPPC by themselves induced a small, PTX-sensitive motile response. However, stimulation of cell motility by LPA, which by itself was also PTX-sensitive, was blocked by SPP and SPPC. In contrast, motility stimulation by thrombin, which by itself was PTX-insensitive, was strongly augmented by the sphingolipids in a PTX-sensitive manner. The bidirectional regulation of LPA- and thrombin-stimulated motility was not due to selective alterations in the activation of Rho GTPases which control cell motility. In fact, RhoA activation and Rho-dependent actin stress fiber formation induced by LPA and thrombin were mimicked, but not altered by SPP and SPPC. We conclude that J82 cells express sphingolipid receptors, coupled via G proteins to several signaling pathways. Most importantly, these sphingolipid receptors potently regulate thrombin- and LPA-stimulated motility, but in opposite directions, suggesting that migration of these human bladder carcinoma cells is controlled by a complex network of interacting extracellular ligands.


Asunto(s)
Carcinoma/fisiopatología , Movimiento Celular/efectos de los fármacos , Lisofosfolípidos/antagonistas & inhibidores , Receptores de Superficie Celular/fisiología , Transducción de Señal/fisiología , Esfingolípidos/metabolismo , Trombina/farmacología , Neoplasias de la Vejiga Urinaria/fisiopatología , Actinas/biosíntesis , Calcio/metabolismo , División Celular/fisiología , AMP Cíclico/metabolismo , Proteínas de Unión al GTP/biosíntesis , Humanos , Lisofosfolípidos/farmacología , Fosfolipasa D/metabolismo , Fosforilcolina/análogos & derivados , Fosforilcolina/farmacología , ARN Mensajero/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esfingosina/análogos & derivados , Esfingosina/farmacología , Células Tumorales Cultivadas , Fosfolipasas de Tipo C/metabolismo
20.
FEBS Lett ; 461(3): 217-22, 1999 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-10567700

RESUMEN

Contribution of sphingosine kinase (SPK)-catalyzed production of sphingosine-1-phosphate (SPP), in comparison to phospholipase C (PLC), to Ca(2+) signalling by epidermal growth factor (EGF) was studied in two HEK-293 cell clones (HEK2 and HEK3), expressing functional EGF receptors and exhibiting release of stored Ca(2+) by intracellular SPP. In HEK3 cells, EGF increased [Ca(2+)](i) and stimulated both, SPK and PLC. [Ca(2+)](i) increase, but not PLC stimulation, was strongly reduced by SPK inhibition. In HEK2 cells, EGF similarly stimulated PLC, but did not increase [Ca(2+)](i) or stimulate SPK, suggesting that intracellular SPP production plays a major role for Ca(2+) signalling by EGF in HEK-293 cells.


Asunto(s)
Señalización del Calcio/fisiología , Receptores ErbB/fisiología , Lisofosfolípidos , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Señalización del Calcio/efectos de los fármacos , Línea Celular , Activación Enzimática , Factor de Crecimiento Epidérmico/farmacología , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Riñón , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fosfatidilinositol Diacilglicerol-Liasa , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Receptores Muscarínicos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Fosfolipasas de Tipo C/metabolismo , Factores de Virulencia de Bordetella/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA