Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
2.
Am J Physiol Regul Integr Comp Physiol ; 304(1): R10-22, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23152113

RESUMO

Acute renal failure (ARF) is a serious medical complication characterized by an abrupt and sustained decline in renal function. Despite significant advances in supportive care, there is currently no effective treatment to restore renal function. PGE(2) is a lipid hormone mediator abundantly produced in the kidney, where it acts locally to regulate renal function; several studies suggest that modulating EP(4) receptor activity could improve renal function following kidney injury. An optimized peptidomimetic ligand of EP(4) receptor, THG213.29, was tested for its efficacy to improve renal function (glomerular filtration rate, renal plasma flow, and urine output) and histological changes in a model of ARF induced by either cisplatin or renal artery occlusion in Sprague-Dawley rats. THG213.29 modulated PGE(2)-binding dissociation kinetics, indicative of an allosteric binding mode. Consistently, THG213.29 antagonized EP(4)-mediated relaxation of piglet saphenous vein rings, partially inhibited EP(4)-mediated cAMP production, but did not affect Gα(i) activation or ß-arrestin recruitment. In vivo, THG213.29 significantly improved renal function and histological changes in cisplatin- and renal artery occlusion-induced ARF models. THG213.29 increased mRNA expression of heme-oxygenase 1, Bcl2, and FGF-2 in renal cortex; correspondingly, in EP(4)-transfected HEK293 cells, THG213.29 augmented FGF-2 and abrogated EP(4)-dependent overexpression of inflammatory IL-6 and of apoptotic death domain-associated protein and BCL2-associated agonist of cell death. Our results demonstrate that THG213.29 represents a novel class of diuretic agent with noncompetitive allosteric modulator effects on EP(4) receptor, resulting in improved renal function and integrity following acute renal failure.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Rim/efeitos dos fármacos , Rim/fisiologia , Oligopeptídeos/uso terapêutico , Receptores de Prostaglandina E Subtipo EP4/agonistas , Recuperação de Função Fisiológica/efeitos dos fármacos , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/patologia , Animais , Cisplatino/efeitos adversos , AMP Cíclico/biossíntese , Modelos Animais de Doenças , Cães , Feminino , Fator 2 de Crescimento de Fibroblastos/biossíntese , Taxa de Filtração Glomerular/efeitos dos fármacos , Células HEK293 , Heme Oxigenase-1/biossíntese , Humanos , Interleucina-6/biossíntese , Masculino , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Ratos , Ratos Sprague-Dawley , Fluxo Plasmático Renal/fisiologia , Veia Safena/efeitos dos fármacos , Veia Safena/patologia , Suínos/fisiologia
3.
Invest Ophthalmol Vis Sci ; 43(10): 3327-37, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12356842

RESUMO

PURPOSE: To test whether platelet-activating factor (PAF) directly causes retinovascular endothelial cell (EC) death. METHODS: Retinovascular density was calculated in rat pups exposed to 80% O(2) from postnatal days (P)6 to P14 (to produce oxygen-induced retinopathy [OIR]), using the adenosine diphosphatase (ADPase) technique, in animals treated with distinct PAF receptor blockers (PCA-4248, BN52021, or THG315). PAF levels were then measured in the retinas. Viability of ECs from piglets and humans in response to C-PAF (a stable PAF analogue) was determined by the reduction of the tetrazolium salt 3-(4,5-dimethyl thiazol-2yl)-2,5-diphenyl tetrazolium bromide (MTT) by viable cells, incorporation of propidium iodide (PI), TUNEL assay, and release of lactate dehydrogenase. Release of thromboxane (TX) was measured in the cell media. RESULTS: PAF levels in retina were markedly increased by exposure of isolated rat retinas to H(2)O(2) (1 micro M) and of rat pups placed in 80% O(2). Exposure to 80% O(2) induced retinal vasoobliteration, which was equally significantly inhibited ( approximately 60%) by all PAF receptor blockers tested. C-PAF increased incorporation of PI by isolated rat retinal microvasculature. Also, C-PAF caused time- and concentration-dependent death of cultured retinal ECs, which was prevented by the PAF receptor antagonist CV-3988. This effect of C-PAF was selective on retinal and neurovascular ECs, but not on other ECs. DNA fragmentation (TUNEL) was hardly detected, and inhibition of apoptosis-related processes by nicotinamide, cyclosporin A, and Z-DEVD-FMK and Z-VAD-FMK (caspase inhibitors) barely protected against death in EC, whereas C-PAF increased release of lactate dehydrogenase, implying that necrosis is the nature of EC death. Finally, C-PAF-induced cell death was preceded by an increase in TXB(2) levels and was prevented by TXA(2) synthase inhibition (with CGS12970). CONCLUSIONS: The data suggest PAF plays a major role in vasoobliteration in OIR by triggering death of neuroretinal microvascular ECs. The cell death seems to be mediated at least in part by TXA(2). These effects of PAF may participate in ischemic retinopathies such as diabetes and retinopathy of prematurity.


Assuntos
Oxigênio , Fator de Ativação de Plaquetas/uso terapêutico , Receptores Acoplados a Proteínas G , Doenças Retinianas/induzido quimicamente , Doenças Retinianas/tratamento farmacológico , Vasos Retinianos/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Humanos , Hiperóxia/complicações , Técnicas In Vitro , Injeções , Microcirculação/efeitos dos fármacos , Estresse Oxidativo , Pericitos/efeitos dos fármacos , Fator de Ativação de Plaquetas/análogos & derivados , Fator de Ativação de Plaquetas/antagonistas & inibidores , Fator de Ativação de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Receptores de Superfície Celular/antagonistas & inibidores , Doenças Retinianas/metabolismo , Doenças Retinianas/patologia , Vasos Retinianos/patologia , Vasos Retinianos/fisiopatologia , Suínos , Tromboxano A2/fisiologia , Corpo Vítreo
4.
Semin Perinatol ; 26(6): 389-97, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12537309

RESUMO

PGF2alpha is an important smooth muscle contractile agent that exerts significant effects on myometrium and is implicated in labor. THG113 was recently identified as a PGF2alpha receptor (FP) antagonist. We characterized the specificity and selectivity of THG113, tested its effects on PGF2alpha-induced smooth muscle contraction, and assessed its efficacy in a model of endotoxin (LPS)-induced preterm labor. [125I]THG113 bound specifically to FP-expressing but not to native (not expressing FP) HEK293 cells. In FP-expressing HEK293 cells, THG113 markedly reduced PGF2alpha-elicited phosphoinositide hydrolysis (IC50 27 nM). Similarly, PGF2alpha-evoked microvascular (retinal) contraction was noncompetitively blocked (by > 90%) by THG113. In contrast, contraction to agonists of homologous prostanoid receptors EP1 and TP (17-phenyl-trinor PGE2 and U46619) was unaffected (< 1%) by high concentrations of THG113 (100 micromol/L); THG113 (100 micromol/L) also did not affect contraction to numerous other agents including platelet activating factor, endothelin, and angiotensin II. Force and duration of PGF2alpha-evoked contractions of myometrial strips of pig (non-pregnant, luteal phase) and mouse (immediately postpartum) were markedly reduced by THG113. In an endotoxin-induced preterm mouse model, lipopolysaccharide (50 microg intraperitioneal) injection at 16 days' gestation resulted in 100% delivery within 15 h; in contrast, 70% of those treated with THG113 (1 mg/day) delivered > 24 h later (at 18 days' gestation; term: 19 days). In addition, in mice injected with lipopolysaccharide and treated 6 h later with THG113 (0.1 mg bolus followed by 1 mg/day) 40% delivered > 48 h later. Fetuses of pregnant mice treated with THG113 were born alive, had higher birth weights (1.6 +/- 0.1 v 1.4 +/- 0.05 g), and appeared healthy. This study describes an effective and selective noncompetitive FP antagonist, THG113, which significantly delays preterm delivery; this provides the basis for future investigations for its use in tocolysis.


Assuntos
Trabalho de Parto Prematuro/prevenção & controle , Receptores de Prostaglandina/antagonistas & inibidores , Tocolíticos/farmacologia , Contração Uterina/efeitos dos fármacos , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Animais , Células Cultivadas , Dinoprosta/metabolismo , Modelos Animais de Doenças , Interações Medicamentosas , Feminino , Humanos , Técnicas In Vitro , Fosfatos de Inositol/biossíntese , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Miométrio/efeitos dos fármacos , Miométrio/metabolismo , Miométrio/fisiologia , Gravidez , Receptores de Prostaglandina/fisiologia , Suínos , Contração Uterina/fisiologia , Vasoconstritores/farmacologia
5.
Am J Physiol Regul Integr Comp Physiol ; 292(3): R1174-83, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17122328

RESUMO

Oxidant stress plays a significant role in hypoxic-ischemic injury to the susceptible microvascular endothelial cells. During oxidant stress, lysophosphatidic acid (LPA) concentrations increase. We explored whether LPA caused cytotoxicity to neuromicrovascular cells and the potential mechanisms thereof. LPA caused a dose-dependent death of porcine cerebral microvascular as well as human umbilical vein endothelial cells; cell death appeared oncotic rather than apoptotic. LPA-induced cell death was mediated via LPA(1) receptor, because the specific LPA(1) receptor antagonist THG1603 fully abrogated LPA's effects. LPA decreased intracellular GSH levels and induced a p38 MAPK/JNK-dependent inducible nitric oxide synthase (NOS) expression. Pretreatment with the antioxidant GSH precursor N-acetyl-cysteine (NAC), as well as with inhibitors of NOS [N(omega)-nitro-l-arginine (l-NNA); 1400W], significantly prevented LPA-induced endothelial cell death (in vitro) to comparable extents; as expected, p38 MAPK (SB203580) and JNK (SP-600125) inhibitors also diminished cell death. LPA did not increase indexes of oxidation (isoprostanes, hydroperoxides, and protein nitration) but did augment protein nitrosylation. Endothelial cytotoxicity by LPA in vitro was reproduced ex vivo in brain and in vivo in retina; THG1603, NAC, l-NNA, and combined SB-203580 and SP600125 prevented the microvascular rarefaction. Data implicate novel properties for LPA as a modulator of the cell redox environment, which partakes in endothelial cell death and ensued neuromicrovascular rarefaction.


Assuntos
Astrócitos/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Oxirredução/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Morte Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Células Endoteliais/ultraestrutura , Endotélio Vascular/citologia , Cinética , Modelos Biológicos , Ratos , Ratos Sprague-Dawley , Sus scrofa
6.
Can J Physiol Pharmacol ; 84(3-4): 377-91, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16902584

RESUMO

Prostaglandins (PGs), platelet-activating factor (PAF), and lysophosphatidic acid (LPA) are ubiquitous lipid mediators that play important roles in inflammation, cardiovascular homeostasis, and immunity and are also known to modulate gene expression of specific pro-inflammatory genes. The mechanism of action of these lipids is thought to be primarily dependent on their specific plasma membrane receptors belonging to the superfamily of G-protein-coupled receptors (GPCR). Increasing evidence suggests the existence of a functional intracellular GPCR population. It has been proposed that immediate effects are mediated via cell surface receptors whereas long-term responses are dependent upon intracellular receptor effects. Indeed, receptors for PAF, LPA, and PGE(2) (specifically EP(1), EP(3), and EP(4)) localize at the cell nucleus of cerebral microvascular endothelial cells of newborn pigs, rat hepatocytes, and cells overexpressing each receptor. Stimulation of isolated nuclei with these lipids reveals biological functions including transcriptional regulation of major genes, namely c-fos, cylooxygenase-2, and endothelial as well as inducible nitric oxide synthase. In the present review, we shall focus on the nuclear localization and signaling of GPCRs recognizing PGE(2), PAF, and LPA phospholipids as ligands. Mechanisms on how nuclear PGE2, PAF, and LPA receptors activate gene transcription and nuclear localization pathways are presented. Intracrine signaling for lipid mediators uncover novel pathways to elicit their effects; accordingly, intracellular GPCRs constitute a distinctive mode of action for gene regulation.


Assuntos
Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Receptores de Prostaglandina E/metabolismo , Animais , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Dinoprostona/metabolismo , Humanos , Lisofosfolipídeos/metabolismo , Fator de Ativação de Plaquetas/metabolismo , Transdução de Sinais
7.
J Biol Chem ; 281(23): 16058-67, 2006 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-16574649

RESUMO

Stimulation of freshly isolated rat hepatocytes with lysophosphatidic acid (LPA) resulted in LPA1 receptor-mediated and nitricoxide-dependent up-regulation of the immediate early genes iNOS (inducible nitric-oxide synthase (NOS)) and mPGES-1 (microsomal prostaglandin E synthase-1). Because LPA is a ligand for both cell surface and intracellular receptor sites and a potent endothelial NOS (eNOS) activator, we hypothesized that NO derived from activated nuclearized eNOS might participate in gene regulation. Herein we show, by confocal microscopy performed on porcine cerebral endothelial cells expressing native LPA1-receptor and eNOS and on HTC4 rat hepatoma cells co-transfected with recombinant human LPA1-receptor and fused eNOS-GFP cDNA, a dynamic eNOS translocation from peripheral to nuclear regions upon stimulation with LPA. Nuclear localization of eNOS and its downstream effector, soluble guanylate cyclase, were demonstrated in situ in rat liver specimens by immunogold labeling using specific antibodies. Stimulation of this nuclear fraction with LPA and the NO donor sodium nitroprusside resulted, respectively, in increased production of nitrite (and eNOS phosphorylation) and cGMP; these separate responses were also correspondingly blocked by NOS inhibitor L-NAME and soluble guanylate cyclase inhibitor ODQ. In addition, sodium nitroprusside evoked a sequential increase in nuclear Ca2+ transients, activation of p42 MAPK, NF-kappaB binding to DNA consensus sequence, and dependent iNOS RNA. This study describes a hitherto unrecognized molecular mechanism by which nuclear eNOS through ensuing NO modulates nuclear calcium homeostasis involved in gene transcription-associated events. Moreover, our findings strongly support the concept of the nucleus as an autonomous signaling compartment.


Assuntos
Genes Precoces , Inflamação/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico/metabolismo , Prostaglandina-Endoperóxido Sintases/genética , Transdução de Sinais , Animais , Sequência de Bases , Núcleo Celular/metabolismo , Células Cultivadas , Primers do DNA , Expressão Gênica/efeitos dos fármacos , Guanilato Ciclase , Humanos , Fígado/metabolismo , Lisofosfolipídeos/farmacologia , Microscopia Confocal , Óxido Nítrico Sintase Tipo III/genética , Prostaglandina-E Sintases , Ratos , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Ácidos Lisofosfatídicos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Guanilil Ciclase Solúvel , Suínos
8.
Am J Obstet Gynecol ; 193(1): 256-66, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16021088

RESUMO

OBJECTIVE: A novel prostaglandin F2alpha receptor antagonist, THG113.31, was tested for the suppression of uterine contractility and delay of preterm labor in sheep. STUDY DESIGN: We determined the tocolytic effectiveness of THG113.31 on contractions that were stimulated in vitro by prostaglandin F2alpha and E2 in longitudinal and circular myometrial strips. We also tested the ability of THG113.31 in vivo to lower uterine electromyographic activity that was induced by the progesterone receptor blocker, RU486, and to delay preterm birth. RESULTS: THG113.31 suppressed the amplitude of prostaglandin F2alpha, but not prostaglandin E2-induced contractions of both circular and longitudinal myometrium (P<.01). The times to delivery after RU486 were 34.8+/-1.1 hours (saline solution) and 41.9+/-0.5 hours (THG113.31; P<.001) or an average delay of 7.1 hours. There were no changes in fetal blood gases (PaO2 , PaCO2 , pH, or SaO2) because of THG113.31. Fetal cortisol levels rose in each group, and fetal and maternal prostaglandin E2 and F2alpha metabolite concentrations rose similarly in both groups. CONCLUSION: THG113.31 specifically suppresses prostaglandin F2alpha-induced myometrial contractility and delays delivery.


Assuntos
Peptídeos/farmacologia , Prenhez/efeitos dos fármacos , Nascimento Prematuro/prevenção & controle , Receptores de Prostaglandina/antagonistas & inibidores , Tocolíticos/farmacologia , Animais , Parto Obstétrico , Dinoprosta/metabolismo , Dinoprosta/farmacologia , Dinoprostona/sangue , Dinoprostona/metabolismo , Dinoprostona/farmacologia , Eletromiografia , Feminino , Sangue Fetal , Hidrocortisona/sangue , Técnicas In Vitro , Mifepristona/farmacologia , Miométrio/efeitos dos fármacos , Miométrio/metabolismo , Miométrio/fisiologia , Gravidez , Prenhez/sangue , Receptores de Progesterona/antagonistas & inibidores , Ovinos , Fatores de Tempo , Contração Uterina/efeitos dos fármacos , Útero/fisiologia
9.
Am J Physiol Regul Integr Comp Physiol ; 286(5): R903-9, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14715488

RESUMO

The synthesis of PGE(2), the major vasodilator prostanoid of the ductus arteriosus (DA), is catalyzed by PGE(2) synthases (PGES). The factors implicated in increased PGE(2) synthesis in the perinatal DA are not known. We studied the developmental changes of PGES along with that of cyclooxygenase (COX)-2 and cytosolic phospholipase A(2) (cPLA(2)) in the DA of fetal (75-90% gestation) and immediately postnatal newborn (NB) piglets. Levels of microsomal PGES (mPGES), COX-2, and PGE(2) in the DA of NB were approximately 7-fold higher than in fetus; activities of cytosolic PGES (cPGES) and cPLA(2) in DA of the fetus and NB did not differ. Because platelet-activating factor (PAF) could regulate COX-2 expression, the former was measured and found to be more abundant in the DA of the NB than of fetus. PAF elicited an increase in mPGES, COX-2, and PGE(2) in fetal DA to levels approaching those of the NB; cPGES, cPLA(2), and COX-1 were unaffected. In perinatal NB DA, PAF receptor antagonists BN-52021 and THG-315 reduced mPGES, COX-2, and PGE(2) levels and were associated with increased DA tone. It is concluded that PAF contributes in regulating DA tone by governing mPGES, COX-2, and ensuing PGE(2) levels in the perinate.


Assuntos
Dinoprostona/metabolismo , Canal Arterial/enzimologia , Oxirredutases Intramoleculares/metabolismo , Animais , Animais Recém-Nascidos , Ciclo-Oxigenase 2 , Citosol/enzimologia , Canal Arterial/embriologia , Técnicas In Vitro , Isoenzimas/metabolismo , Fosfolipases A/metabolismo , Fator de Ativação de Plaquetas/metabolismo , Prostaglandina-E Sintases , Prostaglandina-Endoperóxido Sintases/metabolismo , Suínos
10.
Am J Physiol Regul Integr Comp Physiol ; 284(4): R928-35, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12626359

RESUMO

Oxidant stress contributes to the pathogenesis of hypoxic-ischemic encephalopathies. Platelet-activating factor (PAF) is generated during oxidant stress. We studied the vasomotor mode of actions of PAF on periventricular (PV) microvessels of fetal ( approximately 75% of term), newborn (1-3 days), and adult pigs. PAF constricted PV microvessels from fetal (29.27 +/- 2.6%) and newborn (22.14 +/- 3.2%) pigs but was ineffective in adults (<2.5%). Specific [(3)H]PAF binding was greater in fetus and newborn than in adults; a concordant developmental PAF-induced inositol phosphate formation was observed. PAF-induced vasoconstriction was abrogated by thromboxane A(2) (TXA(2)) synthase and receptor inhibitors, calcium channel blockers, and by removal of endothelium; vasoconstriction to TXA(2) mimetic U-46619 did not differ with age. Immunoreactive TXA(2) synthase expression and PAF-evoked TXA(2) formation revealed a fetus> newborn>adult profile. Thus the greater PAF-induced PV microvascular constriction in younger subjects seems attributable to greater PAF receptor density and mostly secondary to TXA(2) formation from endothelium. The resulting decrease in blood flow may contribute to the increased vulnerability of the PV brain regions to oxidant stress-induced injury in immature subjects.


Assuntos
Envelhecimento/fisiologia , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Fator de Ativação de Plaquetas/farmacologia , Vasoconstrição/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Encéfalo/enzimologia , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Feto/efeitos dos fármacos , Feto/fisiologia , Fosfatos de Inositol/metabolismo , Modelos Logísticos , Suínos/fisiologia , Tromboxano B2/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa