Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Exp Pathol ; 103(1): 4-12, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34655121

RESUMO

The process of gastric ulcer healing includes cell migration, proliferation, angiogenesis and re-epithelialization. Platelets contain angiogenesis stimulating factors that induce angiogenesis. Thromboxane A2 (TXA2 ) not only induces platelet activity but also angiogenesis. This study investigated the role of TXA2 in gastric ulcer healing using TXA2 receptor knockout (TPKO) mice. Gastric ulcer healing was suppressed by treatment with the TXA2 synthase inhibitor OKY-046 and the TXA2 receptor antagonist S-1452 compared with vehicle-treated mice. TPKO showed delayed gastric ulcer healing compared with wild-type mice (WT). The number of microvessels and CD31 expression were lower in TPKO than in WT mice, and TPKO suppressed the expression of transforming growth factor beta (TGF-ß) and vascular endothelial growth factor A (VEGF-A) in areas around gastric ulcers. Immunofluorescence assays showed that TGF-ß and VEGF-A co-localized with platelets. Gastric ulcer healing was significantly reduced in WT mice transplanted with TPKO compared with WT bone marrow. These results suggested that TP signalling on platelets facilitates gastric ulcer healing through TGF-ß and VEGF-A.


Assuntos
Neovascularização Patológica/metabolismo , Úlcera Gástrica/tratamento farmacológico , Tromboxanos/farmacologia , Cicatrização/efeitos dos fármacos , Animais , Camundongos Endogâmicos C57BL , Ativação Plaquetária/efeitos dos fármacos , Prostaglandinas/farmacologia , Receptores de Tromboxanos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Úlcera Gástrica/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
2.
Am J Physiol Renal Physiol ; 320(4): F537-F547, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33491563

RESUMO

Local or systemic inflammation can severely impair urinary bladder functions and contribute to the development of voiding disorders in millions of people worldwide. Isoprostanes are inflammatory lipid mediators that are upregulated in the blood and urine by oxidative stress and may potentially induce detrusor overactivity. The aim of the present study was to investigate the effects and signal transduction of isoprostanes in human and murine urinary bladders in order to provide potential pharmacological targets in detrusor overactivity. Contraction force was measured with a myograph in murine and human urinary bladder smooth muscle (UBSM) ex vivo. Isoprostane 8-iso-PGE2 and 8-iso-PGF2α evoked dose-dependent contraction in the murine UBSM, which was abolished in mice deficient in the thromboxane prostanoid (TP) receptor. The responses remained unaltered after removal of the mucosa or incubation with tetrodotoxin. Smooth muscle-specific deletion of Gα12/13 protein or inhibition of Rho kinase by Y-27632 decreased the contractions. In Gαq/11-knockout mice, responses were reduced and in the presence of Y-27632 abolished completely. In human UBSM, the TP agonist U-46619 evoked dose-dependent contractions. Neither atropine nor the purinergic receptor antagonist pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid decreased the effect, indicating that TP receptors directly mediate detrusor muscle contraction. 8-iso-PGE2 and 8-iso-PGF2α evoked dose-dependent contraction in the human UBSM, and these responses were abolished by the TP antagonist SQ-29548 and were decreased by Y-27632. Our results indicate that isoprostanes evoke contraction in murine and human urinary bladders, an effect mediated by the TP receptor. The G12/13-Rho-Rho kinase pathway plays a significant role in mediating the contraction and therefore may be a potential therapeutic target in detrusor overactivity.NEW & NOTEWORTHY Voiding disorders affect millions of people worldwide. Inflammation can impair urinary bladder functions and contribute to the development of detrusor overactivity. The effects and signal transduction of inflammatory lipid mediator isoprostanes were studied in human and murine urinary bladders ex vivo. We found that isoprostanes evoke contraction, an effect mediated by thromboxane prostanoid receptors. The G12/13-Rho-Rho kinase signaling pathway plays a significant role in mediating the contraction and therefore may be a potential therapeutic target.


Assuntos
Isoprostanos/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Antagonistas de Prostaglandina/farmacologia , Receptores de Prostaglandina/efeitos dos fármacos , Receptores de Tromboxanos/efeitos dos fármacos , Animais , Humanos , Prostaglandinas/farmacologia , Receptores de Tromboxanos/fisiologia
3.
FASEB J ; 34(2): 2568-2578, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31908041

RESUMO

Although recognized to have an in vivo vasodepressor effect blunted by the vasoconstrictor effect of E-prostanoid receptor-3 (EP3), prostaglandin E2 (PGE2 ) evokes contractions of many vascular beds that are sensitive to antagonizing the thromboxane prostanoid receptor (TP). This study aimed to determine the direct effect of PGE2 on renal arteries and/or the whole renal vasculature and how each of these two receptors is involved in the responses. Experiments were performed on isolated vessels and perfused kidneys of wild-type mice and/or mice with deficiency in TP (TP-/- ), EP3 (EP3-/- ), or both TP and EP3 (TP-/- /EP3-/- ). Here we show that PGE2 (0.001-30 µM) evoked not only contraction of main renal arteries, but also a decrease of flow in perfused kidneys. EP3-/- diminished the response to 0.001-0.3 µM PGE2 , while TP-/- reduced that to the prostanoid of higher concentrations. In TP-/- /EP3-/- vessels and perfused kidneys, PGE2 did not evoke contraction but instead resulted in vasodilator responses. These results demonstrate that PGE2 functions as an overall direct vasoconstrictor of the mouse renal vasculature with an effect reflecting the vasoconstrictor activities outweighing that of dilation. Also, our results suggest that EP3 dominates the vasoconstrictor effect of PGE2 of low concentrations (≤0.001-0.3 µM), but its effect is further added by that of TP, which has a higher efficacy, although activated by higher concentrations (from 0.01 µM) of the same prostanoid PGE2 .


Assuntos
Dinoprostona/farmacologia , Receptores de Prostaglandina E Subtipo EP3/efeitos dos fármacos , Receptores de Tromboxanos/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Animais , Dinoprosta/farmacologia , Rim/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Prostaglandinas/farmacologia , Receptores de Prostaglandina/efeitos dos fármacos , Tromboxanos/farmacologia , Vasoconstrição/fisiologia , Vasoconstritores/farmacologia
4.
Crit Care ; 23(1): 42, 2019 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-30744667

RESUMO

BACKGROUND: Several lipid metabolites in cerebrospinal fluid are correlated with poor outcomes in aneurysmal subarachnoid haemorrhage. Most of these metabolites bind to ubiquitous thromboxane-prostaglandin (TP) receptors, causing vasoconstriction and inflammation. Here, we evaluated terutroban (TBN), a specific TP receptor antagonist, for the prevention of post-haemorrhage blood-brain barrier disruption, neuronal apoptosis and delayed cerebral hypoperfusion. METHODS: The rat double subarachnoid haemorrhage model was produced by twice injecting (days 1 and 2) autologous blood into the cisterna magna. Seventy-eight male Sprague-Dawley rats were assigned to experimental groups. Rats exposed to subarachnoid haemorrhage were allocated to no treatment (SAH group) or TBN treatment by gastric gavage during the first 5 days after haemorrhage (SAH+TBN group). Control rats received artificial cerebrospinal fluid injections (CSF group). Sham-operated rats with or without TBN administration were also studied. Body weight and Garcia neurological scores were assessed on day 2 and day 5. We used nanoscale single-photon emission computed tomography (nanoSPECT) to measure brain uptake of three radiolabelled agents: 99mTechnetium-diethylenetriaminepentacetate (99mTc-DTPA), which indicated blood-brain barrier permeability on day 3, 99mTechnetium-annexin V-128 (99mTc-Anx-V128), which indicated apoptosis on day 4, and 99mTechnetium-hexamethylpropyleneamineoxime (99mTc-HMPAO), which indicated cerebral perfusion on day 5. Basilar artery narrowing was verified histologically, and cerebral TP receptor agonists were quantified. RESULTS: 99mTc-DTPA uptake unveiled blood-brain barrier disruption in the SAH group. TBN mitigated this disruption in the brainstem area. 99mTc-Anx-V128 uptake was increased in the SAH group and TBN diminished this effect in the cerebellum. 99mTc-HMPAO uptake revealed a global decreased perfusion on day 5 in the SAH group that was significantly counteracted by TBN. TBN also mitigated basilar artery vasoconstriction, neurological deficits (on day 2), body weight loss (on day 5) and cerebral production of vasoconstrictors such as Thromboxane B2 and Prostaglandin F2α. CONCLUSIONS: Based on in vivo nanoscale imaging, we demonstrated that TBN protected against blood-brain barrier disruption, exerted an anti-apoptotic effect and improved cerebral perfusion. Thus, TP receptor antagonists showed promising results in treating post-haemorrhage neurovascular events.


Assuntos
Naftalenos/farmacocinética , Propionatos/farmacocinética , Receptores de Tromboxanos/efeitos dos fármacos , Hemorragia Subaracnóidea/tratamento farmacológico , Animais , Artéria Basilar/patologia , Masculino , Naftalenos/farmacologia , Naftalenos/uso terapêutico , Desempenho Físico Funcional , Propionatos/farmacologia , Propionatos/uso terapêutico , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/prevenção & controle , Resultado do Tratamento
5.
Am J Physiol Cell Physiol ; 307(6): C571-9, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25080488

RESUMO

A multitude of environmental signaling molecules influence monocyte and macrophage innate and adaptive immune responses, including ATP and prostanoids. Interestingly, purinergic (P2) and eicosanoid receptor signaling interact such that the activation of P2 receptors leads to prostanoid production, which can then interfere with P2Y-mediated macrophage migration. Recent studies suggest that blockade of 5-lipoxygenase (5-LOX) in macrophages can activate a permeation pathway involved in the influx of dye and the release of ATP. Here, we provide evidence that pannexin1 (Panx1) is a component of this pathway and present the intracellular signaling molecules linking the thromboxane (TP) receptor to Panx1-mediated dye influx and ATP release. Using pharmacological tools and transgenic mice deficient in Panx1, we show that two 5-LOX pathway inhibitors induce ATP release and influx of dye in a Panx1-dependent manner. Electrophysiological recordings performed in wild-type and Panx1-deficient macrophages confirmed that these 5-LOX pathway inhibitors activate currents characteristic of Panx1 channels. We found that the mechanism by which Panx1 channels are activated under this condition involves activation of the TP receptor that is mediated by the cAMP/PKA pathway. This is to our knowledge the first evidence for the involvement of Panx1 in the TP receptor signaling pathway. Future studies aimed to clarify the contribution of this TP-Panx1 signaling network to macrophage immune responses are likely to be important for targeting inflammatory and autoimmune diseases.


Assuntos
Conexinas/metabolismo , Inibidores de Lipoxigenase/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Receptores de Tromboxanos/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Conexinas/deficiência , Conexinas/genética , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Macrófagos Peritoneais/metabolismo , Masculino , Potenciais da Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Receptores de Tromboxanos/metabolismo , Transdução de Sinais/efeitos dos fármacos
6.
Am J Physiol Renal Physiol ; 305(6): F830-8, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23884143

RESUMO

The present renal hemodynamic study tested the hypothesis that CD38 and superoxide anion (O2(·-)) participate in the vasoconstriction produced by activation of thromboxane prostanoid (TP) receptors in the mouse kidney. CD38 is the major mammalian ADP-ribosyl cyclase contributing to vasomotor tone through the generation of cADP-ribose, a second messenger that activates ryanodine receptors to release Ca(2+) from the sarcoplasmic reticulum in vascular smooth muscle cells. We evaluated whether the stable thromboxane mimetic U-46619 causes less pronounced renal vasoconstriction in CD38-deficient mice and the involvement of O2(·-) in U-46619-induced renal vasoconstriction. Our results indicate that U-46619 activation of TP receptors causes renal vasoconstriction in part by activating cADP-ribose signaling in renal resistance arterioles. Based on maximal renal blood flow and renal vascular resistance responses to bolus injections of U-46619, CD38 contributes 30-40% of the TP receptor-induced vasoconstriction. We also found that the antioxidant SOD mimetic tempol attenuated the magnitude of vasoconstriction by U-46619 in both groups of mice, suggesting mediation by O2(·-). The degree of tempol blockage of U-46619-induced renal vasoconstriction was greater in wild-type mice, attenuating renal vasoconstriction by 40% compared with 30% in CD38-null mice. In other experiments, U-46619 rapidly stimulated O2(·-) production (dihydroethidium fluorescence) in isolated mouse afferent arterioles, an effect abolished by tempol. These observations provide the first in vivo demonstration of CD38 and O2(·-) involvement in the vasoconstrictor effects of TP receptor activation in the kidney and in vitro evidence for TP receptor stimulation of O2(·-) production by the afferent arteriole.


Assuntos
ADP-Ribosil Ciclase 1/fisiologia , Rim/irrigação sanguínea , Glicoproteínas de Membrana/fisiologia , Superóxidos/farmacologia , Vasoconstrição/efeitos dos fármacos , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , ADP-Ribosil Ciclase 1/deficiência , Animais , Arteríolas/efeitos dos fármacos , Óxidos N-Cíclicos/farmacologia , Rim/metabolismo , Glicoproteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Tromboxanos/efeitos dos fármacos , Receptores de Tromboxanos/fisiologia , Marcadores de Spin
7.
Arterioscler Thromb Vasc Biol ; 32(10): 2468-74, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22879580

RESUMO

OBJECTIVE: Thromboxane A(2) and prostacyclin are thromboregulatory prostaglandins. The inflammatory C-reactive protein (CRP) promotes thrombosis after vascular injury, presumably via potentiation of thromboxane activity. Using a genetic approach, we investigated the role of thromboxane receptor (TP) pathway in CRP-induced thrombosis. METHODS AND RESULTS: Four genetically engineered mice strains were used: C57BL/6 wild-type, human CRP transgenic (CRPtg), thromboxane receptor-deficient (Tp(-/-)), and CRPtgTp(-/-) mice. CRP and TP expression were correlated, and suppression of CRP expression using small interfering RNA/CRP led to reduction in TP expression. Platelet-endothelial adherence was increased in CRPtg and suppressed in CRPtgTP(-/-)and CRPtg cells that were suppressed with TP small interfering RNA. TP deficiency in both platelets and endothelial cells was synergistic in affecting platelet-endothelial interactions. Time until arterial occlusion, measured after photochemical injury, was significantly shorter in CRPtg and prolonged in CRPtgTp(-/-) compared with controls (n=10-15, 35±3.4, 136±13.8, and 67±8.9 minutes, respectively; P<0.05). CONCLUSIONS: TP pathway is of major importance in CRP-induced thrombosis. The expression of TP is increased in CRPtg endothelial cells, and its blockade significantly suppresses the prothrombotic effect of CRP.


Assuntos
Proteína C-Reativa/fisiologia , Receptores de Tromboxanos/fisiologia , Transdução de Sinais/fisiologia , Trombose/fisiopatologia , Adulto , Animais , Plaquetas/patologia , Plaquetas/fisiologia , Proteína C-Reativa/deficiência , Proteína C-Reativa/genética , Adesão Celular/fisiologia , Modelos Animais de Doenças , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , RNA Interferente Pequeno/farmacologia , Receptores de Tromboxanos/deficiência , Receptores de Tromboxanos/efeitos dos fármacos , Trombose/patologia , Transfecção
8.
Exp Physiol ; 97(7): 895-904, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22447972

RESUMO

This study aimed to determine whether cyclo-oxygenase-1 (COX-1) mediates dilatation of mouse arteries via synthesis of prostacyclin (PGI(2)) and, if so, how PGI(2) (IP) receptors contribute and whether thromboxane prostanoid (TP) receptors are implicated in the process. Mesenteric arteries were isolated from wild-type mice or mice with COX-1 deficiency (COX-1(-/-)). The vasomotor reaction to the COX substrate arachidonic acid (AA) was determined with isometric force measurement, while the in vitro production or the plasma level of the PGI(2) metabolite 6-keto-PGF(1α) was analysed with high-performance liquid chromatography-mass spectroscopy or enzyme immunoassay, respectively. Results showed that AA, which evoked endothelium-dependent 6-keto-PGF(1α) production, elicited relaxation that was inhibited or enhanced by antagonizing IP or TP receptors, respectively. Also, IP receptor blockade resulted in contraction in response to AA (following NO synthase inhibition), which was prevented by a concomitant TP receptor antagonism. Meanwhile, COX-1(-/-) or COX-1 inhibition abolished the in vitro 6-keto-PGF(1α) production and reduced the relaxation or contraction observed with AA. Real-time PCR showed that whereas TP receptor mRNAs were detected at similar levels, IP receptor mRNAs were present at higher levels in the branches than in the main stem of the mesenteric artery. In addition, antagonizing the IP receptors enhanced the contraction evoked by PGI(2) in the carotid artery. Also, we noted that COX-1(-/-) mice had a reduced basal plasma 6-keto-PGF(1α) level. These results demonstrate an explicit vasodilator role for COX-1-mediated endothelial PGI(2) synthesis and suggest that the functionally opposing IP and TP receptors concomitantly mediate the vasomotor reaction to PGI(2), with the dilator activity of IP receptors being compromised by the vasoconstrictor effect of TP receptors and vice versa.


Assuntos
Ácido Araquidônico/farmacologia , Receptores de Prostaglandina/metabolismo , Receptores de Tromboxanos/metabolismo , 6-Cetoprostaglandina F1 alfa/metabolismo , Animais , Ácido Araquidônico/metabolismo , Ciclo-Oxigenase 1/deficiência , Ciclo-Oxigenase 1/metabolismo , Epoprostenol/biossíntese , Epoprostenol/metabolismo , Masculino , Artérias Mesentéricas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Epoprostenol/efeitos dos fármacos , Receptores de Prostaglandina/efeitos dos fármacos , Receptores de Tromboxanos/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos
9.
Life Sci ; 286: 120073, 2021 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-34688694

RESUMO

AIMS: Thromboxane (TxA2) is synthesized from arachidonic acid (AA) via thromboxane synthase (TxS) enzyme and induces vasoconstriction via TP receptor. Our aim is to compare the effects of aspirin, TxS inhibitor and TP receptor antagonist on vascular reactivity of bypass grafts (saphenous vein and internal mammary artery). MAIN METHODS: Using isolated organ bath, saphenous vein and internal mammary artery preparations were incubated with TP receptor antagonist, TxS inhibitor, aspirin, IP or EP4 receptor antagonist. Then prostaglandin (PG)E2, PGF2α, phenylephrine and AA were administered in concentration-dependent manner. The expression of prostanoid receptor and PGI2 synthase (PGIS) enzyme was determined by Western Blot. KEY FINDINGS: TP receptor antagonist inhibited the contraction induced by PGE2, PGF2α, and AA but not that induced by phenylephrine in both types of vessels. Aspirin increased phenylephrine-induced contraction only in internal mammary artery and decreased AA-induced contraction in saphenous vein. TxS inhibitor decreased both PGE2 and AA-induced contraction in both types of vessels. This decrease was reversed by co-incubation of TxS inhibitor and IP/EP4 receptor antagonists. The expressions of EP3 receptor and PGIS enzyme were greater in internal mammary artery compared to saphenous vein while IP and TP receptors expressed at similar levels. SIGNIFICANCE: TP receptor antagonist and TxS inhibitor are more effective to reduce contraction induced by different spasmogens in comparison to aspirin. Our results suggest that TP receptor antagonist and TxS inhibitor might have an advantage over aspirin due to their preventive effect on increased vascular reactivity observed in post-operative period of coronary artery bypass grafting.


Assuntos
Artéria Torácica Interna/efeitos dos fármacos , Veia Safena/efeitos dos fármacos , Ácido Araquidônico/metabolismo , Aspirina/farmacologia , Benzofuranos/farmacologia , Carbazóis/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Masculino , Artéria Torácica Interna/metabolismo , Músculo Liso Vascular/metabolismo , Fenilefrina/farmacologia , Receptores de Prostaglandina/metabolismo , Receptores de Tromboxanos/antagonistas & inibidores , Receptores de Tromboxanos/efeitos dos fármacos , Receptores de Tromboxanos/metabolismo , Veia Safena/metabolismo , Sulfonamidas/farmacologia , Tromboxano A2/farmacologia , Tromboxano-A Sintase/antagonistas & inibidores , Tromboxano-A Sintase/efeitos dos fármacos , Tromboxano-A Sintase/metabolismo , Tromboxanos/antagonistas & inibidores , Tromboxanos/metabolismo , Vasoconstrição/efeitos dos fármacos
10.
J Pharmacol Exp Ther ; 328(1): 231-9, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18836067

RESUMO

Epoxy- and dihydroxy-eicosatrienoic acids (EETs and DHETs) are vasoactive cytochrome P450 metabolites of arachidonic acid. Interestingly, however, the mechanism(s) by which EETs/DHETs mediate smooth muscle relaxation remains unclear. In contrast to previous reports, where dilation was purportedly large-conductance Ca(2+)-activated K(+) (BK(Ca)) and/or transient receptor potential cation channel, subfamily V, member 4 (TRPV4) channel-mediated, 14,15-EET-induced vasodilation [reversal of contractile tone established with the thromboxane receptor (TP) agonist 15-hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic acid (U-46619)] was unaltered in BK(Ca) and TRPV4 knockout mouse isolated aortae compared with wild-type controls, indicating a significant BK(Ca)/TRPV4-resistant mechanism. Whereas all EET and DHET regioisomers reversed U-46619 contraction in rat aortae and mouse mesenteric resistance arteries, these eicosanoids failed to alter phenylephrine-induced contraction, suggesting that they mediated dilation via a "TP-selective" mechanism. Competitive TP antagonism was also observed in nonvascular tissue, including rat fundus and tertiary bronchus, indicating that the effect is not specific to blood vessels. Such effects were TP-selective because 14,15-EET failed to inhibit "non-TP" prostanoid receptor-mediated function in multiple cell/tissue-based assays (K(b) > 10 microM). In accordance, 14,15-EET inhibited specific [(3)H]7-(3-((2-((phenylamino)carbonyl)hydrazino)-methyl)-7-oxabicyclo(2.2.1)hept-2-yl)-5-heptenoic acid (SQ-29548) binding to human recombinant TP receptor, with a K(i) value of 3.2 microM, and it showed weaker affinity for non-TP prostanoid receptors, including DP, FP, EP(1-4), and IP receptors (K(i) values of 6.1, 5.3, 42.6, 19.7, 13.2, 20.2, and >25 microM, respectively) and no appreciable affinity (K(i) values >10 microM) for a diverse array of pharmacologically distinct receptors, including the leukotriene receptors Cys-LT(1/2) and BLT(1). As such, EETs/DHETs represent a unique class of "endogenous" G protein-coupled receptor competitive antagonists, inducing vasodilation via direct TP inhibition. Thus, EETs/DHETs represent novel autoregulatory agents, directly modulating the actions of cyclooxygenase-derived eicosanoids following arachidonic acid mobilization.


Assuntos
Ácido 8,11,14-Eicosatrienoico/farmacologia , Aorta Torácica/fisiologia , Vasodilatação/fisiologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Animais , Aorta Torácica/efeitos dos fármacos , Ácidos Araquidônicos/farmacologia , Cobaias , Ácidos Hidroxieicosatetraenoicos/farmacologia , Masculino , Camundongos , Camundongos Knockout , Ratos , Ratos Sprague-Dawley , Receptores de Tromboxanos/agonistas , Receptores de Tromboxanos/efeitos dos fármacos , Receptores de Tromboxanos/fisiologia , Circulação Esplâncnica/efeitos dos fármacos , Circulação Esplâncnica/fisiologia , Canais de Cátion TRPV/deficiência , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/fisiologia , Traqueia/efeitos dos fármacos , Traqueia/fisiologia , Resistência Vascular/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia , Vasodilatação/efeitos dos fármacos
11.
Thromb Haemost ; 100(1): 83-9, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18612542

RESUMO

Aspirin 'resistance' (AR) is a phenomenon of uncertain etiology describing decreased platelet inhibition by aspirin. We studied whether (i) platelets in AR demonstrate increased basal sensitivity to a lower degree of stimulation and (ii) platelet aggregation with submaximal stimulation could predict responses to aspirin. Serum thromboxane B(2) (TxB(2)) levels and platelet aggregation with light transmission aggregometry (LTA) were measured at baseline and 24 hours after 325 mg aspirin administration in 58 healthy subjects. AR was defined as the upper sixth of LTA (> or = 12%) to 1.5 mM AA. Baseline platelet aggregation with submaximal concentrations of agonists [ADP 2 microM, arachidonic acid (AA) 0.75 mM, collagen 0.375 and 0.5 microg/ml] was greater in AR subjects compared with non-AR subjects, but not with higher concentrations (ADP 5 microM and 20 microM, AA 1.5 mM and collagen 1 microg/ml). Post-aspirin platelet aggregation was elevated in AR subjects with both submaximal and maximal stimulation. Baseline and post-aspirin serum TxB(2) were higher in AR subjects and decreased further with ex-vivo COX-1 inhibition, suggesting incompletely suppressed COX-1 activity. Pre-aspirin platelet aggregation to 0.75 AA demonstrated a dichotomous response with 29/58 subjects having aggregation < or = 15% and 29/58 subjects having aggregation > or = 75%. In the high aggregation group 28% had AR compared to 6% in the non-AR group (p = 0.04). In conclusion, platelets in AR subjects demonstrate increased basal sensitivity to submaximal stimulation, which could predict responses to antiplatelet therapy.


Assuntos
Ácido Araquidônico , Aspirina/farmacologia , Plaquetas/efeitos dos fármacos , Resistência a Medicamentos , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Difosfato de Adenosina , Adulto , Aspirina/uso terapêutico , Plaquetas/metabolismo , Colágeno , Ciclo-Oxigenase 1/sangue , Inibidores de Ciclo-Oxigenase/farmacologia , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Inibidores da Agregação Plaquetária/uso terapêutico , Testes de Função Plaquetária , Valor Preditivo dos Testes , Receptores de Tromboxanos/sangue , Receptores de Tromboxanos/efeitos dos fármacos , Valores de Referência , Tromboxano B2/sangue , Falha de Tratamento
12.
Eur J Pharmacol ; 592(1-3): 81-6, 2008 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-18647601

RESUMO

Neuromedin U is a hypothalamic peptide involved in energy homeostasis and stress responses. The peptide, when administered intracerebroventricularly (i.c.v.), decreases food intake and body weight while increasing body temperature and heat production. We examined the effect of i.c.v. administered neuromedin U on plasma catecholamines with regard to the brain prostanoid using anesthetized rats. Neuromedin U (0.1, 0.5 and 1 nmol/animal, i.c.v.) effectively elevated plasma adrenaline (a maximal response was obtained at 0.5 nmol/animal), but had little effect on plasma noradrenaline. However, intravenously administered neuromedin U (0.5 nmol/animal) had no effect on plasma catecholamines. Neuromedin U (0.5 nmol/animal, i.c.v.)-induced elevation of plasma adrenaline was effectively reduced by intracerebroventricular pretreatments with indomethacin (an inhibitor of cyclooxygenase) (0.6 and 1.2 micromol/animal), furegrelate (an inhibitor of thromboxane A2 synthase) (0.9 and 1.8 micromol/animal) and (+)-S-145 (a blocker of prostanoid TP receptors) (250 and 625 nmol/animal), respectively. The neuromedin U-induced adrenaline response was also abolished by acute bilateral adrenalectomy. These results suggest that centrally administered neuromedin U evokes the secretion of adrenaline from the adrenal medulla by brain prostanoid TP receptor-mediated mechanisms in rats.


Assuntos
Epinefrina/sangue , Neuropeptídeos/farmacologia , Receptores de Tromboxanos/efeitos dos fármacos , Anestesia , Anestésicos Inalatórios , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Benzofuranos/farmacologia , Química Encefálica/efeitos dos fármacos , Compostos Bicíclicos com Pontes/farmacologia , Catecolaminas/metabolismo , Inibidores Enzimáticos/farmacologia , Ácidos Graxos Monoinsaturados/farmacologia , Indometacina/farmacologia , Injeções Intraventriculares , Masculino , Neuropeptídeos/administração & dosagem , Antagonistas de Prostaglandina/farmacologia , Ratos , Ratos Wistar , Tromboxano-A Sintase/antagonistas & inibidores , Uretana
13.
Vasc Health Risk Manag ; 4(3): 637-45, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18827913

RESUMO

A common pathophysiological course in vascular diseases is an overwhelming activation and aggregation of blood platelets, which results in atherothrombosis. By causing the last decisive step of cerebral, coronary, or peripheral arterial ischemia thrombotic complications of atherosclerotic disease represent a major player in death cause statistics of most western countries. The development of novel therapies against platelet-dependent thrombosis and the concurrent improvement of existing therapeutic strategies thus is a paramount focus of pharmaceutical research. Currently, efficiency, dosing and indications of established antiplatelet substances are being re-evaluated, whilst new, so far unrecognized molecular targets for inhibition of platelet activity come up front. This not only allows for interesting new therapeutical options, but also widens our insight into the role platelets play in atherosclerosis in general. This article summarizes the relevant pathophysiology of platelet activation, presents current concepts in antiplatelet drug therapy, and highlights the role of platelets in vascular diseases apart from atherothrombosis.


Assuntos
Aterosclerose/tratamento farmacológico , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Trombose/tratamento farmacológico , Aterosclerose/fisiopatologia , Clopidogrel , Stents Farmacológicos , Humanos , Piperazinas/administração & dosagem , Ativação Plaquetária/fisiologia , Agregação Plaquetária/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Cloridrato de Prasugrel , Antagonistas do Receptor Purinérgico P2 , Piridinas/uso terapêutico , Receptores de Tromboxanos/efeitos dos fármacos , Transdução de Sinais/fisiologia , Tiofenos/administração & dosagem , Trombose/fisiopatologia , Ticlopidina/administração & dosagem , Ticlopidina/análogos & derivados , Ticlopidina/uso terapêutico
14.
Atherosclerosis ; 191(2): 250-8, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16797553

RESUMO

Sanguinarine is a plant alkaloid present in the root of Sanguinaria canadensis and Poppy fumaria species. Sanguinarine has been used as an antiseptic mouth rinse and a toothpaste additive to reduce dental plaque and gingival inflammation. In this study, we investigated the antiplatelet effects of sanguinarine, aiming to extend its potential pharmacological applications. Sanguinarine inhibited platelet aggregation induced by arachidonic acid (AA), collagen, U46619 and sub-threshold concentration of thrombin (0.05 U/ml) with IC(50) concentrations of 8.3, 7.7, 8.6 and 4.4 microM, respectively. Sanguinarine (5-10 microM) inhibited 10-31% of platelet TXB(2) production, but not platelet aggregation induced by higher concentration of thrombin (0.1 U/ml). SQ29548, a thromboxane receptor antagonist, inhibited the AA-induced platelet aggregation but not TXB(2) production. Sanguinarine suppressed cyclooxygenase-1 (COX-1) activity (IC(50)=28 microM), whereas its effect on COX-2 activity was minimal. Sanguinarine (8, 10 microM) further inhibited the AA-induced Ca(2+) mobilization by 27-62%. In addition, SQ22536, an adenylate cyclase inhibitor, attenuated the inhibitory effect of sanguinarine toward AA-induced platelet Ca(2+) mobilization and aggregation. These results suggest that sanguinarine is a potent antiplatelet agent, which activates adenylate cyclase, inhibits platelet Ca(2+) mobilization, TXB(2) production as well as suppresses COX-1 enzyme activity. Sanguinarine may have therapeutic potential for treatment of cardiovascular diseases related to platelet aggregation.


Assuntos
Alcaloides/farmacologia , Benzofenantridinas/farmacologia , Plaquetas/efeitos dos fármacos , Cálcio/metabolismo , AMP Cíclico/metabolismo , Inibidores de Ciclo-Oxigenase/farmacologia , Isoquinolinas/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Tromboxano B2/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Adenilil Ciclases/metabolismo , Animais , Ácido Araquidônico/farmacologia , Plaquetas/metabolismo , Colágeno/farmacologia , Ciclo-Oxigenase 1/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Técnicas In Vitro , Coelhos , Receptores de Tromboxanos/efeitos dos fármacos , Receptores de Tromboxanos/metabolismo , Trombina/farmacologia
15.
Eur J Pharmacol ; 562(3): 227-35, 2007 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-17362923

RESUMO

Considering the potential physiological, pharmacological and therapeutic relevance of synergistic interaction of thromboxane A(2) with adrenaline at postjunctional receptor sites, we examined whether sub-threshold concentrations of thromboxane A(2) mimetic U-46619 (9,11-dideoxy-9alpha, 11alpha-methanoepoxy prostaglandin F(2alpha)) could amplify adrenaline-induced contraction in human umbilical vein. The receptor involved in U-46619-induced potentiation of adrenaline contractility was also investigated. Umbilical cords (n=125) from healthy patients after full-term vaginal or caesarean deliveries were employed. The vein was dissected out of cords and rings used for isolated organ bath experiments or reverse transcription-polymerase chain reaction (RT-PCR) and Western blot. Presence of endothelium did not modify U-46619-induced contraction in human umbilical vein. Prostanoid TP-selective receptor antagonist, SQ-29548 (7-[3-[[2-[(phenylamino)carbonyl]hydrazino]methyl]-7-oxabicyclo[2.2.1]hept-2-yl]-[1S(1alpha,2alpha(Z),3alpha,4alpha)]-5-Heptenoic acid), inhibited U-46619-induced contraction (pA(2)=8.22+/-0.11). U-46619 sub-threshold concentrations (0.1-0.3 nM) potentiated adrenaline-vasoconstriction response in a concentration-dependent manner. SQ-29548 (0.1 microM) abolished this potentiation. Using RT-PCR, we found that human umbilical vein rings with or without endothelium express the prostanoid TP(alpha), but not the prostanoid TP(beta) receptor isoform. Western blot allowed the identification of proteins with an electrophoretic mobility (47- and 55-kDa) indistinguishable from human platelet prostanoid TP receptor, a rich source of prostanoid TP(alpha) receptor isoform. Collectively, present results demonstrate that prostanoid TP(alpha) is the major receptor isoform localized on smooth muscle cells which participate in both direct vasoconstriction and potentiating effects of U-46619 on adrenaline contractions in human umbilical vein. These results suggest that thromboxane A(2) may interact synergistically with adrenaline in pathophysiological situations that lead to an increase of its umbilical venous levels (e.g. preeclampsia associated with fetal distress) raising the possibility of vasoconstriction affecting fetal blood flow.


Assuntos
Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Epinefrina/farmacologia , Receptores de Tromboxanos/efeitos dos fármacos , Veias Umbilicais/efeitos dos fármacos , Vasoconstritores/farmacologia , Western Blotting , Compostos Bicíclicos Heterocíclicos com Pontes , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Endotélio Vascular/metabolismo , Ácidos Graxos Insaturados , Feminino , Humanos , Hidrazinas , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Isoformas de Proteínas/metabolismo , Receptores de Tromboxanos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Veias Umbilicais/metabolismo , Vasoconstrição/efeitos dos fármacos
17.
Oncogene ; 22(29): 4488-97, 2003 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-12881705

RESUMO

Trefoil peptides (TFFs) are now considered as scatter factors, proinvasive and angiogenic agents acting through cyclooxygenase-2 (COX-2)- and thromboxane A2 receptor (TXA2-R)-dependent signaling pathways. As expression and activation levels of the epidermal growth factor receptor (EGFR) predict the metastatic potential of human colorectal cancers, the purpose of this study was to establish whether the EGF receptor tyrosine kinase (EGFR-TK) contributes to cellular invasion induced by TFFs in kidney and colonic cancer cells. Both the dominant negative form of the EGFR (HER-CD533) and the EGFR-TK inhibitor ZD1839 (Iressa) abrogated cellular invasion induced by pS2, spasmolytic polypeptide (SP) and the src oncogene, but not by ITF and the TXA2-R. Similarly, EGFR-TK inhibition by ZD1839 reversed the invasive phenotype promoted by the constitutively activated form of the EGFR (EGFRvIII) and the EGFR agonists transforming growth factor alpha (TGFalpha), amphiregulin and EGF. We also provide evidence that TFFs, EGFRvIII, and TGFalpha trigger common proinvasive pathways using the PI3'-kinase and Rho/Rho- kinase cascades. These findings identify the EGFR-TK as a key signaling element for pS2- and SP-mediated cellular invasion. It is concluded that although pS2, SP and ITF belong to the same family of inflammation- and cancer-associated regulatory peptides, they do not control identical signaling networks.


Assuntos
Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Receptores ErbB/metabolismo , Substâncias de Crescimento/farmacologia , Rim/metabolismo , Mucinas , Proteínas Musculares , Neuropeptídeos , Peptídeos/farmacologia , Proteínas , Anfirregulina , Animais , Células Cultivadas , Neoplasias do Colo/tratamento farmacológico , Cães , Família de Proteínas EGF , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/efeitos dos fármacos , Receptores ErbB/genética , Gefitinibe , Genes src/genética , Glicoproteínas/metabolismo , Glicoproteínas/farmacologia , Substâncias de Crescimento/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Rim/citologia , Rim/efeitos dos fármacos , Mutação , Invasividade Neoplásica , Peptídeos/metabolismo , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Quinazolinas/farmacologia , Receptores de Tromboxanos/antagonistas & inibidores , Receptores de Tromboxanos/efeitos dos fármacos , Receptores de Tromboxanos/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador alfa/metabolismo , Fator de Crescimento Transformador alfa/farmacologia , Fator Trefoil-1 , Fator Trefoil-2 , Fator Trefoil-3 , Proteínas Supressoras de Tumor
18.
Biochim Biophys Acta ; 1181(2): 148-54, 1993 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-8481403

RESUMO

We have characterized the thromboxane (TX) A2/prostaglandin (PG) H2 receptor in glomeruli isolated from the rat using the agonist radioligand [125I]-BOP. Binding of [125I]-BOP was highly specific, stereoselective, and to a single class of high affinity binding sites (Kd = 1.16 +/- 0.22 nM and Bmax = 348 +/- 32 fmol/mg protein; n = 6). Binding of [125I]-BOP was competed for by the agonist ONO11113 (Kd = 50.8 +/- 8.0 nM; n = 4) and the antagonists SQ29548 (Kd = 15.8 +/- 1.0 nM; n = 3), L657925 (Kd = 12.1 +/- 2.2 nM; n = 3) and L657926 (Kd = 1642 +/- 135 nM; n = 3). I-BOP also produced a TXA2/PGH2 receptor-mediated rise in [Ca2+]i in isolated glomeruli In adriamycin-induced nephrotic syndrome in the rat, the development of proteinuria is reported to be dependent on increased renal TXA2 production. We therefore examined whether or not changes in glomerular TXA2/PGH2 receptors occur between control and nephrotic rats. No changes in expression or affinity of either glomerular or platelet TXA2/PGH2 receptors were observed. Kd and Bmax values for isolated glomeruli were 1.45 +/- 0.24 nM and 406 +/- 72 fmol/mg for controls and 1.22 +/- 0.25 nM and 321 +/- 62 fmol/mg for nephrotic rats (n = 6).


Assuntos
Compostos Bicíclicos Heterocíclicos com Pontes , Doxorrubicina/toxicidade , Glomérulos Renais/metabolismo , Síndrome Nefrótica/metabolismo , Receptores de Prostaglandina/metabolismo , Receptores de Tromboxanos/metabolismo , Animais , Ligação Competitiva , Compostos Bicíclicos com Pontes/farmacologia , Ácidos Graxos Insaturados/farmacologia , Cinética , Masculino , Síndrome Nefrótica/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina/antagonistas & inibidores , Receptores de Prostaglandina/efeitos dos fármacos , Receptores de Tromboxanos/antagonistas & inibidores , Receptores de Tromboxanos/efeitos dos fármacos , Receptores de Tromboxano A2 e Prostaglandina H2 , Tromboxano A2/análogos & derivados , Tromboxano A2/farmacologia
19.
Biochim Biophys Acta ; 1221(1): 83-8, 1994 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-8130280

RESUMO

Thromboxane A2 (TXA2) and its precursor prostaglandin H2 (PGH2) stimulate platelet activation through interaction with TXA2/PGH2 receptors. We and others have shown that these receptors undergo homologous desensitization upon prolonged exposure to thromboxane A2 mimetics. Phosphorylation of receptors has previously been reported to be an important mechanism for receptor desensitization. In the present study we examined the possibility that homologous desensitization of human platelet TXA2/PGH2 receptors may involve phosphorylation. The ATP pool of human platelets was metabolically prelabeled with 32Pi and the labeled platelets were subsequently exposed for 1 and 10 min to the stable TXA2/PGH2 mimetic, U46619 (1 microM). TXA2/PGH2 receptors were purified approx. 2000-fold by affinity and wheatgerm lectin chromatography and subjected to SDS-PAGE followed by autoradiography. A phosphorylated plasma membrane glycoprotein (M(r) = 50-57 kDa) was detected with characteristics similar to the TXA2/PGH2 receptor. This glycoprotein was found to be phosphorylated in the unstimulated state but phosphorylation was increased by exposure to U46619. Phosphorylation occurred rapidly and was inhibited when platelets were preincubated with the TXA2/PGH2 receptor antagonist, SQ29548 (5 microM), before being stimulated with U46619. These results suggest that human platelet TXA2/PGH2 receptors are phosphoproteins and that the level of phosphorylation is increased during homologous desensitization.


Assuntos
Plaquetas/metabolismo , Receptores de Prostaglandina/metabolismo , Receptores de Tromboxanos/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico , Plaquetas/efeitos dos fármacos , Compostos Bicíclicos Heterocíclicos com Pontes , Membrana Celular/metabolismo , Cromatografia de Afinidade , Ácidos Graxos Insaturados , Humanos , Hidrazinas/farmacologia , Técnicas In Vitro , Cinética , Fosfatos/sangue , Radioisótopos de Fósforo , Fosforilação , Endoperóxidos Sintéticos de Prostaglandinas/farmacologia , Prostaglandina H2 , Prostaglandinas H/sangue , Ensaio Radioligante , Receptores de Prostaglandina/efeitos dos fármacos , Receptores de Prostaglandina/isolamento & purificação , Receptores de Tromboxanos/efeitos dos fármacos , Receptores de Tromboxanos/isolamento & purificação , Receptores de Tromboxano A2 e Prostaglandina H2 , Tromboxano A2/análogos & derivados , Tromboxano A2/antagonistas & inibidores , Tromboxano A2/farmacologia , Vasoconstritores/farmacologia
20.
Hypertension ; 29(1 Pt 2): 303-9, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9039119

RESUMO

We recently identified a subgroup of rabbits (called nonresponders) that were deficient in vascular thromboxane A2 receptors. Thromboxane A2-mediated platelet aggregation was not different between responders and nonresponders. In the present study, we utilized these nonresponders as a model to study the relative contribution of the platelet and vascular thromboxane A2 receptors to the observed hemodynamic responses associated with arachidonic acid-induced sudden death. Mean arterial pressure was slightly but not significantly lower in the nonresponders compared with the responders. However, nonresponders were protected from arachidonic acid-induced sudden death. While 100% of the responders died at the 2.0 mg dose of arachidonic acid, only 27% of nonresponders died at this same dose. Administration of the thromboxane A2 mimetic U46619 (5 micrograms/kg IV) decreased blood pressure by 41 +/- 6 mm Hg in responders but had no effect in the nonresponders. The affinity and density of thromboxane A2 receptors in cultured aortic vascular smooth muscle cells obtained from both responders and nonresponders were assessed using radioligand binding. The Kd values were not different (4.4 +/- 1.0 versus 2.4 +/- 0.6 nmol/L, responder versus nonresponder). However, there was a significant decrease in the density of receptors from vascular smooth muscle cells of nonresponders (Bmax = 397 +/- 59 versus 157 +/- 59 fmol/10(6) cells, responder versus nonresponder, P < .01). U46619 produced a concentration-dependent increase in [3H]-thymidine incorporation into responder vascular smooth muscle cells but had no effect in the nonresponder cells. Using an anti-thromboxane A2 receptor antibody, we compared the amount of receptor expressed in aortic tissue obtained from responders and nonresponders. Consistent with the results observed with [3H]-thymidine uptake and radioligand binding assays, the expression of thromboxane A2 receptor protein was decreased in nonresponder compared with responder vascular tissue. Platelet thromboxane A2 receptor expression was not different. These studies demonstrate that the vascular smooth muscle cells of nonresponder rabbits are deficient in the thromboxane A2 receptor. Furthermore, the reduction in arachidonic acid-induced sudden death in nonresponders indicates that the vascular smooth muscle thromboxane A2 receptor mediates this effect.


Assuntos
Ácido Araquidônico , Morte Súbita , Músculo Liso Vascular/química , Endoperóxidos Sintéticos de Prostaglandinas/farmacologia , Receptores de Tromboxanos/fisiologia , Tromboxano A2/análogos & derivados , Vasoconstritores/farmacologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico , Animais , Aorta/química , Aorta/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Western Blotting , Frequência Cardíaca/efeitos dos fármacos , Masculino , Coelhos , Receptores de Tromboxanos/deficiência , Receptores de Tromboxanos/efeitos dos fármacos , Tromboxano A2/sangue , Tromboxano A2/farmacologia , Timidina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA