Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Thromb Haemost ; 14(4): 815-27, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26806373

RESUMO

BACKGROUND: Acquired and inherited bleeding disorders may present in the neonatal period with devastating lifelong effects. Diagnosing bleeding disorders in the neonatal population could aid in preventing and treating the associated complications. However, currently available platelet function testing is limited in neonates, owing to difficulties in obtaining an adequate blood volume, a lack of normal reference ranges, and an incomplete understanding of the neonatal platelet functional phenotype. OBJECTIVE: To develop small-volume, whole blood platelet function assays in order to quantify and compare neonatal and adult platelet function. METHODS AND RESULTS: Peripheral blood was obtained from healthy, full-term neonates at 24 h of life. Platelet activation, secretion and aggregation were measured via flow cytometry. Platelet adhesion and aggregation were assessed under static and flow conditions. As compared with adult platelets, peripheral neonatal platelet P-selectin expression and integrin glycoprotein IIbIIIa activation were significantly reduced in response to the G-protein-coupled receptor (GPCR) agonists thrombin receptor activator peptide-6 (TRAP-6), ADP, and U46619, and the immunoreceptor tyrosine-based activation motif (ITAM) signaling pathway agonists collagen-related peptide (CRP) and rhodocytin. Neonatal platelet aggregation was markedly reduced in response to TRAP-6, ADP, U46619, CRP and rhodocytin as compared with adult platelets. The extents of neonatal and adult platelet adhesion and aggregate formation under static and shear conditions on collagen and von Willebrand factor were similar. CONCLUSIONS: As compared with adult platelets, we found that neonatal platelet activation and secretion were blunted in response to GPCR or ITAM agonists, whereas the extent of neonatal platelet adhesion and aggregate formation was similar to that of adult platelets.


Assuntos
Plaquetas/citologia , Ativação Plaquetária , Adesividade Plaquetária , Agregação Plaquetária , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/química , Difosfato de Adenosina/química , Adulto , Proteína C-Reativa/química , Separação Celular , Citometria de Fluxo , Glicoproteínas/química , Hemorragia/sangue , Humanos , Motivo de Ativação do Imunorreceptor Baseado em Tirosina , Recém-Nascido , Lectinas Tipo C/química , Oligopeptídeos/química , Testes de Função Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/química , Complexo Glicoproteico GPIb-IX de Plaquetas/química , Receptores Acoplados a Proteínas G/agonistas , Transdução de Sinais
2.
Thromb Res ; 136(6): 1310-7, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26553017

RESUMO

INTRODUCTION: Thromboxane A2 (TXA2) can induce the platelet aggregation and lead to thrombosis. This will cause the low-reflow phenomenon after ischemic stroke and aggravate the damage of brain issues. Therefore, it is potential to develop the drugs inhibiting TXA2 pathway to treat cerebral ischemia. AIM: This study aims to prove the protective effect of N2 (4-(2-(1H-imidazol-1-yl) ethoxy)-3-methoxybenzoic acid) on focal cerebral ischemia and reperfusion injury through platelet aggregation inhibition. MATERIALS AND METHODS: Middle cerebral artery occlusion/reperfusion (MCAO/R) is used as the animal model. Neurological deficit score, Morris water maze, postural reflex test, Limb-use asymmetry test, infarct volume, and water content were performed to evaluate the protective effect of N2 in MCAO/R rats. 9, 11-dieoxy-11α, 9α-methanoepoxyprostaglandin F2α (U46619) or adenosine diphosphate (ADP) was used as the inducer of platelet aggregation. RESULTS AND CONCLUSIONS: N2 can improve the motor function, learning and memory ability in MCAO/R rats while reducing the infarct volume. N2 can inhibit TXA2 formation but promote PGI2, and can inhibit platelet aggregation induced by U46619 and ADP. Further, N2 inhibits thrombosis with a minor adverse effect of bleeding than Clopidogrel. In conclusion, N2 can produce the protective effect on MCAO/R brain injury through inhibiting TXA2 formation, platelet aggregation and thrombosis.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Agregação Plaquetária/efeitos dos fármacos , Acidente Vascular Cerebral/tratamento farmacológico , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/química , Difosfato de Adenosina/química , Animais , Derivação Arteriovenosa Cirúrgica , Coagulação Sanguínea , Encéfalo/patologia , Edema/patologia , Ensaio de Imunoadsorção Enzimática , Epoprostenol/química , Feminino , Imidazóis/química , Masculino , Aprendizagem em Labirinto , Ratos , Ratos Sprague-Dawley , Trombose/fisiopatologia , Tromboxano A2/química , Ácido Vanílico/análogos & derivados , Ácido Vanílico/química
3.
Biomed Res Int ; 2014: 728019, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24868545

RESUMO

Amarogentin, an active principle of Gentiana lutea, possess antitumorigenic, antidiabetic, and antioxidative properties. Activation of platelets is associated with intravascular thrombosis and cardiovascular diseases. The present study examined the effects of amarogentin on platelet activation. Amarogentin treatment (15~60 µM) inhibited platelet aggregation induced by collagen, but not thrombin, arachidonic acid, and U46619. Amarogentin inhibited collagen-induced phosphorylation of phospholipase C (PLC) γ2, protein kinase C (PKC), and mitogen-activated protein kinases (MAPKs). It also inhibits in vivo thrombus formation in mice. In addition, neither the guanylate cyclase inhibitor ODQ nor the adenylate cyclase inhibitor SQ22536 affected the amarogentin-mediated inhibition of platelet aggregation, which suggests that amarogentin does not regulate the levels of cyclic AMP and cyclic GMP. In conclusion, amarogentin prevents platelet activation through the inhibition of PLC γ2-PKC cascade and MAPK pathway. Our findings suggest that amarogentin may offer therapeutic potential for preventing or treating thromboembolic disorders.


Assuntos
Iridoides/química , Sistema de Sinalização das MAP Quinases , Fosfolipase C gama/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Proteína Quinase C/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/química , Adenina/análogos & derivados , Adenina/química , Trifosfato de Adenosina/química , Animais , Ácido Araquidônico/química , Colágeno/química , GMP Cíclico/metabolismo , Guanilato Ciclase/antagonistas & inibidores , Humanos , Camundongos , Oxidiazóis/química , Extratos Vegetais/química , Quinoxalinas/química , Trombina/química , Tromboembolia/tratamento farmacológico , Tromboembolia/prevenção & controle , Trombose/tratamento farmacológico
4.
Chembiochem ; 15(6): 892-9, 2014 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-24623680

RESUMO

CYP5A1 is a membrane-associated cytochrome P450 that metabolizes the cyclooxygenase product prostaglandin (PGH2 ) into thromboxane A2 (TXA2 ), a potent inducer of vasoconstriction and platelet aggregation. Although CYP5A1 is an ER-bound protein, the role of membranes in modulating the thermodynamics and kinetics of substrate binding to this protein has not been elucidated. In this work, we incorporated thromboxane synthase into lipid bilayers of nanodiscs for functional studies. We measured the redox potential of CYP5A1 in nanodiscs and showed that the redox potential is within a similar range of other drug-metabolizing P450 enzymes in membranes. Further, we showed that binding of substrate to CYP5A1 can induce conformational changes in the protein that block small-molecule ligand egress by measuring the kinetics of cyanide binding to CYP5A1 as a function of substrate concentration. Notably, we observed that sensitivity to cyanide binding was different for two substrate analogues, U44069 and U46619, thus indicating that they bind differently to the active site of CYP5A1. We also characterized the effects of the different lipids on CYP5A1 catalytic activity by using nanodiscs to create unary, binary, and ternary lipid systems. CYP5A1 activity increased dramatically in the presence of charged lipids POPS and POPE, as compared to the unary POPC system. These results suggest the importance of lipid composition on modulating the activity of CYP5A1 to increase thromboxane formation.


Assuntos
Bicamadas Lipídicas/metabolismo , Nanoestruturas/química , Tromboxano-A Sintase/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/química , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/metabolismo , Domínio Catalítico , Cianetos/química , Cianetos/metabolismo , Humanos , Cinética , Bicamadas Lipídicas/química , Oxirredução , Endoperóxidos Sintéticos de Prostaglandinas/química , Endoperóxidos Sintéticos de Prostaglandinas/metabolismo , Ligação Proteica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Especificidade por Substrato , Termodinâmica , Tromboxano-A Sintase/química , Tromboxano-A Sintase/genética
5.
FEBS Lett ; 588(6): 962-9, 2014 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-24530534

RESUMO

The hydrophobic cavity of lipocalin-type prostaglandin D synthase (L-PGDS) has been suggested to accommodate various lipophilic ligands through hydrophobic effects, but its energetic origin remains unknown. We characterized 18 buffer-independent binding systems between human L-PGDS and lipophilic ligands using isothermal titration calorimetry. Although the classical hydrophobic effect was mostly detected, all complex formations were driven by favorable enthalpic gains. Gibbs energy changes strongly correlated with the number of hydrogen bond acceptors of ligand. Thus, the broad binding capability of L-PGDS for ligands should be viewed as hydrophilic interactions delicately tuned by enthalpy-entropy compensation using combined effects of hydrophilic and hydrophobic interactions.


Assuntos
Oxirredutases Intramoleculares/química , Lipocalinas/química , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/química , Substituição de Aminoácidos , Bilirrubina/química , Biliverdina/química , Domínio Catalítico , Hormônios Gonadais/química , Hemina/química , Humanos , Interações Hidrofóbicas e Hidrofílicas , Oxirredutases Intramoleculares/genética , Ligantes , Lipocalinas/genética , Naftalenossulfonatos/química , Ligação Proteica , Esteroides/química , Termodinâmica , Hormônios Tireóideos/química , Tretinoína/química
7.
Thromb Haemost ; 109(6): 1120-30, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23426129

RESUMO

Sulforaphane, a dietary isothiocyanate found in cruciferous vegetables, has been shown to exert beneficial effects in animal models of cardiovascular diseases. However, its effect on platelet aggregation, which is a critical factor in arterial thrombosis, is still unclear. In the present study, we show that sulforaphane inhibited human platelet aggregation caused by different receptor agonists, including collagen, U46619 (a thromboxane A2 mimic), protease-activated receptor 1 agonist peptide (PAR1-AP), and an ADP P2Y12 receptor agonist. Moreover, sulforaphane significantly reduced thrombus formation on a collagen-coated surface under whole blood flow conditions. In exploring the underlying mechanism, we found that sulforaphane specifically prevented phosphatidylinositol 3-kinase (PI3K)/Akt signalling, without markedly affecting other signlaling pathways involved in platelet aggregation, such as protein kinase C activation, calcium mobilisation, and protein tyrosine phosphorylation. Although sulforaphane did not directly inhibit the catalytic activity of PI3K, it caused ubiquitination of the regulatory p85 subunit of PI3K, and prevented PI3K translocation to membranes. In addition, sulforaphane caused ubiquitination and degradation of phosphoinositide-dependent kinase 1 (PDK1), which is required for Akt activation. Therefore, sulforaphane is able to inhibit the PI3K/Akt pathway at two distinct sites. In conclusion, we have demonstrated that sulforaphane prevented platelet aggregation and reduced thrombus formation in flow conditions; our data also support that the inhibition of the PI3K/Akt pathway by sulforaphane contributes it antiplatelet effects.


Assuntos
Isotiocianatos/química , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores da Agregação Plaquetária/química , Agregação Plaquetária/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/química , Animais , Plaquetas/citologia , Cálcio/metabolismo , Bovinos , Ativação Enzimática , Humanos , Inibidores da Agregação Plaquetária/farmacologia , Proteína Quinase C/metabolismo , Receptor PAR-1/metabolismo , Transdução de Sinais , Sulfóxidos , Tromboxano A2/química , Tirosina/química , Vasoconstritores/química , Verduras
8.
Yakugaku Zasshi ; 131(11): 1575-81, 2011.
Artigo em Japonês | MEDLINE | ID: mdl-22041695

RESUMO

Lipocalin-type prostaglandin (PG) D synthase (L-PGDS) is a multi functional protein acting as a PGD(2) synthesizing enzyme, a transporter or scavenger of various lipophilic ligands, and an amyloid ß chaperon in the brain. L-PGDS is a member of the lipocalin superfamily and has the ability to bind various lipophilic molecules such as prostanoid, retinoid, bile pigment, and amyloid ß peptide. However, the molecular mechanism for a wide variety of ligand binding has not been well understood. In this study, we determined by NMR the structure of recombinant mouse L-PGDS and L-PGDS/PGH(2) analog complex. L-PGDS has the typical lipocalin fold, consisting of an eight-stranded ß-barrel and a long α-helix. The interior of the barrel formed a hydrophobic cavity opening to the upper end of the barrel, the size of which was larger than those of other lipocalins and the cavity contained two pockets. Kinetic studies and molecular docking studies based on the result of NMR titration experiments provide the direct evidence for two binding sites for PGH(2) and retinoic acid in the large cavity of L-PGDS. Structural comparison of L-PGDS/U-46619 complex with apo-L-PGDS showed that the H2-helix, CD-loop, and EF-loop located at the upper end of the ß-barrel change the conformation to cover the entry of the cavity upon U-46619 binding. These results indicated that the two binding sites in the large cavity and induced fit mechanism were responsible for the broad ligand specificity of L-PGDS.


Assuntos
Oxirredutases Intramoleculares/química , Oxirredutases Intramoleculares/fisiologia , Lipocalinas/química , Lipocalinas/fisiologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/química , Sítios de Ligação , Desenho de Fármacos , Ligantes , Terapia de Alvo Molecular , Complexos Multiproteicos , Ressonância Magnética Nuclear Biomolecular , Prostaglandina H2 , Ligação Proteica , Conformação Proteica , Sono/genética , Sono/fisiologia , Tretinoína
9.
Biochemistry ; 47(2): 680-8, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18081314

RESUMO

High-resolution NMR spectroscopy was used to determine the docking of a substrate (prostaglandin H2) mimic (U46619) to the engineered prostacyclin (PGI2) synthase (PGIS) in solution. The binding of U46619 to the PGIS protein was demonstrated by 1D NMR titration, and the significant perturbation of the chemical shifts of protons at C-11, H2C, and H20 of U46619 were observed upon U46619 binding to the engineered PGIS in a concentration-dependent manner. The detailed conformational change and 3D structure of the PGIS-bound U46619 were further demonstrated by 2D 1H NMR experiments using the transferred NOE technique. The distances between the protons H20 and H2, H18 and H2, and H18 and H4 are shorter following their binding to the PGIS in solution-down to within 5 A. These shorter distances resulted in a widely open conformation, where the triangle shape of the unbound U46619 changed to a more compact conformation with an oval shape. The bound conformation of U46619 fits the crystal structure of the PGIS substrate binding pocket considerably better than that of the unbound U46619. The residues important to the substrate binding in the active site pocket of PGIS were also predicted. For example, Trp282 could be one of the most important residues and is suspected to play a role in the determination of specific catalytic function, which has been established by the docking studies using the NMR structure of the PGIS-bound form of U46619 and the PGIS crystal structure. These studies have provided the structural information for the interaction of the PGIS with its substrate mimic. The noted conformational changes where the C-6 position is closer to the C-9 position of U46619 provided the first experimental data for understanding the molecular mechanism of the catalytic function of PGIS in the isomerization of PGH2 to prostacyclin.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Epoprostenol/biossíntese , Oxirredutases Intramoleculares/metabolismo , Mimetismo Molecular , Mutagênese , Engenharia de Proteínas , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/química , Cristalografia por Raios X , Humanos , Espectroscopia de Ressonância Magnética , Conformação Molecular , Mutação/genética , Prostaglandina H2/química , Prótons , Solubilidade , Soluções , Especificidade por Substrato , Titulometria , Triptofano
10.
Prostaglandins Other Lipid Mediat ; 84(1-2): 14-23, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17643884

RESUMO

Thromboxane A(2) (TXA(2)) and 8-iso-PGF(2alpha) are two prostanoid agonists of the thromboxane A(2) receptor (TP), whose activation has been involved in platelet aggregation and atherosclerosis. Agents able to counteract the actions of these agonists are of great interest in the treatment and prevention of cardiovascular events. Here, we investigated in vitro and in vivo the pharmacological profile of BM-520, a new TP antagonist. In our experiments, this compound showed a great binding affinity for human washed platelets TP receptors, and prevented human platelet activation and aggregation induced by U-46619, arachidonic acid and 8-iso-PGF(2alpha). The TP receptor antagonist property of BM-520 was confirmed by its relaxing effect on rat aorta smooth muscle preparations precontracted with U-46619 and 8-iso-PGF(2alpha). Further, its TP antagonism was also demonstrated in vivo in guinea pig after a single intravenous injection (10 mg kg(-1)). We conclude that this novel TP antagonist could be a promising therapeutic tool in pathologies such as atherosclerosis where an increased production of TXA(2) and 8-iso-PGF(2alpha), as well as TP activation are well-established pathogenic events.


Assuntos
Aorta/metabolismo , Dinoprosta/análogos & derivados , Difenilamina/análogos & derivados , Músculo Liso Vascular/metabolismo , Compostos de Sulfonilureia/química , Tromboxano A2/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/química , Animais , Aorta/efeitos dos fármacos , Plaquetas/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes , Dinoprosta/química , Difenilamina/química , Difenilamina/farmacologia , Ácidos Graxos Insaturados , Cobaias , Humanos , Hidrazinas/química , Masculino , Modelos Químicos , Ratos , Ratos Wistar , Compostos de Sulfonilureia/farmacologia
11.
J Biochem ; 135(4): 455-63, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15115769

RESUMO

Prostacyclin synthase (PGIS), which catalyzes the conversion of prostaglandin (PG) H(2) to prostacyclin (PGI(2)), is a member of the cytochrome P-450 (P450) superfamily, CYP8A1. To study the enzymatic and protein characteristics of human PGIS, the enzyme was overexpressed in Spodoptera frugiperda 21 (Sf21) cells using the baculovirus expression system. PGIS was expressed in the microsomes of the infected Sf21 cells after culture in 5 microg/ml hematin-supplemented medium for 72 h. The holoenzyme was isolated from the solubilized microsomal fraction by calcium phosphate gel absorption and purified to homogeneity by DEAE-Sepharose and hydroxyapatite column chromatography. The K(m) and V(max) values of the purified human PGIS for PGH(2) were 30 microM and 15 micromol/min/mg of protein at 24 degrees C, respectively. The optical absorption and EPR spectra of the enzyme revealed the characteristics of a low-spin form of P450 in the oxidized state. The carbon monoxide-reduced difference spectrum, however, exhibited a peak at 418 nm rather than 450 nm. The addition of a PGH(2) analogue, U46619, to the enzyme produced an oxygen-ligand type of the difference spectrum with maximum absorption at 407 nm and minimum absorption at 430 nm. Treatment with another PGH(2) analogue, U44069, produced a peak at 387 nm and a trough at 432 nm in the spectrum (Type I), while treatment with tranylcypromine, a PGIS inhibitor, produced a peak at 434 nm and a trough at 412 nm (Type II). A Cys441His mutant of the enzyme possessed no heme-binding ability or enzyme activity. Thus, we succeeded in obtaining a sufficient amount of the purified recombinant human PGIS from infected insect cells for spectral analyses that has high specific activity and the characteristics of a P450, indicating substrate specificity.


Assuntos
Sistema Enzimático do Citocromo P-450/química , Oxirredutases Intramoleculares/química , Proteínas Recombinantes/química , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/química , 6-Cetoprostaglandina F1 alfa/química , 6-Cetoprostaglandina F1 alfa/metabolismo , Animais , Baculoviridae/genética , Linhagem Celular , Cromatografia por Troca Iônica , Cromatografia Líquida , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/isolamento & purificação , Durapatita/química , Espectroscopia de Ressonância de Spin Eletrônica , Eletroforese em Gel de Poliacrilamida , Expressão Gênica , Humanos , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/isolamento & purificação , Cinética , Oxirredução , Mutação Puntual , Endoperóxidos Sintéticos de Prostaglandinas/química , Prostaglandina H2/química , Prostaglandina H2/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Espectrofotometria , Spodoptera , Tromboxano A2/análogos & derivados , Transfecção , Tranilcipromina/química
12.
Br J Pharmacol ; 139(1): 156-62, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12746234

RESUMO

1. Alpha(2)-adrenoceptor-mediated contractions in porcine blood vessels can be enhanced in the presence of the thromboxane-mimetic U46619, and forskolin. The aim of this study was to determine the role of U46619 in the enhanced contractions, and to determine whether signalling through the ERK-MAP kinase pathway is involved. 2. Responses to the alpha(2)-adrenoceptor agonist UK14304 (1 micro M) were increased from 22+/-3% of the response to 60 mM KCl to 68+/-12% (n=8, mean+/-s.e.m.) in the presence of a low concentration of U46619 (< 20% of the 60 mM KCl response). 3. Both the direct and the U46619-enhanced UK14304 responses were inhibited by 50 microM PD98059, an inhibitor of the ERK-MAP kinase pathway. UK14304-induced contractions were associated with an increase in ERK2 phosphorylation, indicating an increased activity. In the presence of U46619, there was an enhanced phosphorylation of ERK2. U46619 on its own had no effect on ERK phosphorylation. 4. Both the direct and enhanced UK14304 contractions were inhibited in the absence of extracellular calcium. These conditions also prevented the increase in ERK2 phosphorylation. This indicates a role for calcium influx in the enhanced contractions. 5. In conclusion, this study demonstrates that precontraction with the thromboxane-mimetic U46619 enhances alpha(2)-adrenoceptor-mediated vasoconstriction through the enhancement of the ERK-MAP kinase pathway, and influx of extracellular calcium.


Assuntos
Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Transdução de Sinais/fisiologia , Tromboxanos/química , Vasoconstrição/efeitos dos fármacos , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/química , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Artérias/efeitos dos fármacos , Artérias/enzimologia , Artérias/fisiologia , Tartarato de Brimonidina , Cálcio/metabolismo , Orelha/irrigação sanguínea , Ativação Enzimática , Flavonoides/farmacologia , Técnicas In Vitro , Mimetismo Molecular , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/fisiologia , Fosforilação , Quinoxalinas/farmacologia , Receptores Adrenérgicos alfa 2/fisiologia , Suínos , Vasoconstrição/fisiologia
13.
Biochemistry ; 42(9): 2542-51, 2003 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-12614148

RESUMO

Thromboxane synthase is a hemethiolate enzyme that catalyzes the isomerization of prostaglandin H2 to thromboxane A2. We report the first resonance Raman (RR) spectra of recombinant human thromboxane synthase (TXAS) in both the presence and the absence of substrate analogues U44069 and U46619. The resting enzyme and its U44069 complex are found to have a 6-coordinate, low spin (6c/ls) heme, in agreement with earlier experiments. The U46619-bound enzyme is detected as a 6c/ls heme too, which is in contradiction with a previous conclusion based on absorption difference spectroscopy. Two new vibrations at 368 and 424 cm(-1) are observed upon binding of the substrate analogues in the heme pocket and are assigned to the second propionate and vinyl bending modes, respectively. We interpret the changes in these vibrational modes as the disruption of the protein environment and the hydrogen-bonding network of one of the propionate groups when the substrate analogues enter the heme pocket. We use carbocyclic thromboxane A2 (CTA2) to convert the TXAS heme cofactor to its 5-coordinate, high spin (5c/hs) form, as is confirmed by optical and RR spectroscopy. In this 5c/hs state of the enzyme, the Fe-S stretching frequency is determined at 350 cm(-1) with excitation at 356.4 nm. This assignment is supported by comparison to the spectrum of resting enzyme excited at 356.4 nm and by exciting at different wavelengths. Implications of our findings for substrate binding and the catalytic mechanism of TXAS will be discussed.


Assuntos
Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/química , Endoperóxidos Sintéticos de Prostaglandinas/química , Tromboxano-A Sintase/química , Sítios de Ligação , Ativação Enzimática , Heme/química , Humanos , Ligação de Hidrogênio , Proteínas Ferro-Enxofre/química , Ligantes , Substâncias Macromoleculares , Modelos Moleculares , Proteínas Recombinantes de Fusão/química , Espectrofotometria Ultravioleta/métodos , Análise Espectral Raman/métodos , Especificidade por Substrato
14.
J Biol Chem ; 275(52): 40679-85, 2000 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-11006279

RESUMO

The present studies describe an investigation for the interaction of N-terminal membrane anchor domain of thromboxane A(2) synthase (TXAS) with its substrate analog in a membrane-bound environment using the two-dimensional NMR technique. TXAS and prostaglandin I(2) synthase (PGIS), respectively, convert the same substrate, prostaglandin H(2) (PGH(2)), to thromboxane A(2) and prostaglandin I(2), which have opposite biological functions. Our topology studies have indicated that the N-terminal region of TXAS has a longer N-terminal endoplasmic reticulum (ER) membrane anchor region compared with the same segment proposed for PGIS. The differences in their interaction with the ER membrane may have an important impact to facilitate their common substrate, PGH(2), across the membrane into their active sites from the luminal to the cytoplasmic side of the ER. To test this hypothesis, we first investigated the interaction of the TXAS N-terminal membrane anchor domain with its substrate analog. A synthetic peptide corresponding to the N-terminal membrane anchor domain (residues 1-35) of TXAS, which adopted a stable helical structure and exhibited a membrane anchor function in the membrane-bound environment, was used to interact with a stable PGH(2) analog,. High resolution two-dimensional NMR experiments, NOESY and TOCSY, were performed to solve the solution structures of in a membrane-mimicking environment using dodecylphosphocholine micelles. Different conformations were clearly observed in the presence and absence of the TXAS N-terminal membrane anchor domain. Through combination of the two-dimensional NMR experiments, completed (1)H NMR assignments of were obtained, and the data were used to construct three-dimensional structures of in H(2)O and dodecylphosphocholine micelles, showing the detailed conformation change upon the interaction with the membrane anchor domain. The observation supported the presence of a substrate interaction site in the N-terminal region. The combination of the structural information of and was able to simulate a solution structure of the unstable TXAS and PGIS substrate, PGH(2).


Assuntos
Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/química , Endoperóxidos Sintéticos de Prostaglandinas/química , Tromboxano-A Sintase/química , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/metabolismo , Sítios de Ligação , Espectroscopia de Ressonância Magnética , Conformação Molecular , Endoperóxidos Sintéticos de Prostaglandinas/metabolismo , Relação Estrutura-Atividade , Tromboxano-A Sintase/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA