Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Biol Chem ; 291(52): 26598-26612, 2016 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-27803165

RESUMO

Integrins function as bi-directional signaling transducers that regulate cell-cell and cell-matrix signals across the membrane. A key modulator of integrin activation is talin, a large cytoskeletal protein that exists in an autoinhibited state in quiescent cells. Talin is a large 235-kDa protein composed of an N-terminal 45-kDa FERM (4.1, ezrin-, radixin-, and moesin-related protein) domain, also known as the talin head domain, and a series of helical bundles known as the rod domain. The talin head domain consists of four distinct lobes designated as F0-F3. Integrin binding and activation are mediated through the F3 region, a critically regulated domain in talin. Regulation of the F3 lobe is accomplished through autoinhibition via anti-parallel dimerization. In the anti-parallel dimerization model, the rod domain region of one talin molecule binds to the F3 lobe on an adjacent talin molecule, thus achieving the state of autoinhibition. Platelet functionality requires integrin activation for adherence and thrombus formation, and thus regulation of talin presents a critical node where pharmacological intervention is possible. A major mechanism of integrin activation in platelets is through heterotrimeric G protein signaling regulating hemostasis and thrombosis. Here, we provide evidence that switch region 2 (SR2) of the ubiquitously expressed G protein (Gα13) directly interacts with talin, relieves its state of autoinhibition, and triggers integrin activation. Biochemical analysis of Gα13 shows SR2 binds directly to the F3 lobe of talin's head domain and competes with the rod domain for binding. Intramolecular FRET analysis shows Gα13 can relieve autoinhibition in a cellular milieu. Finally, a myristoylated SR2 peptide shows demonstrable decrease in thrombosis in vivo Altogether, we present a mechanistic basis for the regulation of talin through Gα13.


Assuntos
Plaquetas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Membrana/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Talina/antagonistas & inibidores , Animais , Sítios de Ligação , Adesão Celular , Células Cultivadas , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Humanos , Camundongos , Modelos Moleculares , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Ligação Proteica , Talina/metabolismo , Trombose/metabolismo , Trombose/patologia
2.
J Biol Chem ; 290(41): 25129-39, 2015 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-26292217

RESUMO

Even though GPCR signaling in human platelets is directly involved in hemostasis and thrombus formation, the sequence of events by which G protein activation leads to αIIbß3 integrin activation (inside-out signaling) is not clearly defined. We previously demonstrated that a conformationally sensitive domain of one G protein, i.e. Gα13 switch region 1 (Gα13SR1), can directly participate in the platelet inside-out signaling process. Interestingly however, the dependence on Gα13SR1 signaling was limited to PAR1 receptors, and did not involve signaling through other important platelet GPCRs. Based on the limited scope of this involvement, and the known importance of G13 in hemostasis and thrombosis, the present study examined whether signaling through another switch region of G13, i.e. Gα13 switch region 2 (Gα13SR2) may represent a more global mechanism of platelet activation. Using multiple experimental approaches, our results demonstrate that Gα13SR2 forms a bi-molecular complex with the head domain of talin and thereby promotes ß3 integrin activation. Moreover, additional studies provided evidence that Gα13SR2 is not constitutively associated with talin in unactivated platelets, but becomes bound to talin in response to elevated intraplatelet calcium levels. Collectively, these findings provide evidence for a novel paradigm of inside-out signaling in platelets, whereby ß3 integrin activation involves the direct binding of the talin head domain to the switch region 2 sequence of the Gα13 subunit.


Assuntos
Plaquetas/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/química , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Talina/química , Talina/metabolismo , Sequência de Aminoácidos , Animais , Plaquetas/citologia , Adesão Celular , Humanos , Camundongos , Células NIH 3T3 , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/agonistas , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais
3.
J Biol Chem ; 287(49): 41218-31, 2012 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-23060452

RESUMO

Dematin is a broadly expressed membrane cytoskeletal protein that has been well characterized in erythrocytes and to a lesser extent in non-erythroid cells. However, dematin's function in platelets is not known. Here, we show that dematin is abundantly expressed in both human and mouse platelets. Platelets harvested from the dematin headpiece knock-out (HPKO) mouse model exhibit a striking defect in the mobilization of calcium in response to multiple agonists of platelet activation. The reduced calcium mobilization in HPKO platelets is associated with concomitant inhibition of platelet aggregation and granule secretion. Integrin α(IIb)ß(3) activation in response to agonists is attenuated in the HPKO platelets. The mutant platelets show nearly normal spreading on fibrinogen and an unaltered basal cAMP level; however, the clot retraction was compromised in the mutant mice. Immunofluorescence analysis indicated that dematin is present both at the dense tubular system and plasma membrane fractions of platelets. Proteomic analysis of dematin-associated proteins in human platelets identified inositol 1,4,5-trisphosphate 3-kinase isoform B (IP3KB) as a binding partner, which was confirmed by immunoprecipitation analysis. IP3KB, a dense tubular system protein, is a major regulator of calcium homeostasis. Loss of the dematin headpiece resulted in a decrease of IP3KB at the membrane and increased levels of IP3KB in the cytosol. Collectively, these findings unveil dematin as a novel regulator of internal calcium mobilization in platelets affecting multiple signaling and cytoskeletal functions. Implications of a conserved role of dematin in the regulation of calcium homeostasis in other cell types will be discussed.


Assuntos
Plaquetas/metabolismo , Cálcio/química , Proteínas dos Microfilamentos/química , Animais , Plaquetas/citologia , Sinalização do Cálcio , Doenças Cardiovasculares/metabolismo , Proteínas do Citoesqueleto/metabolismo , Citosol/química , Citosol/metabolismo , Membrana Eritrocítica/metabolismo , Hemostasia , Humanos , Camundongos , Modelos Biológicos , Mutação , Ativação Plaquetária , Estrutura Terciária de Proteína , Transdução de Sinais
4.
Arterioscler Thromb Vasc Biol ; 32(3): 768-77, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22207728

RESUMO

OBJECTIVE: Recent studies have shown a role for Rac1 in regulating platelet functions, but how Rac1 is activated in platelets remains unclear. Phosphatidylinositol-3,4,5-trisphosphate-dependent Rac exchanger 1 (P-Rex1) was originally identified in neutrophils that regulates phagocyte functions. We sought to examine whether P-Rex1 plays a role in platelet activation. METHODS AND RESULTS: Western blotting showed P-Rex1 expression in mouse and human platelets. Mice lacking P-Rex1 exhibited prolonged bleeding time and increased rebleeding. When challenged with low doses of the G protein-coupled receptor (GPCR) agonists U46619 and thrombin, P-Rex1-/- platelets displayed significantly reduced secretion and aggregation compared with wild-type platelets. Increasing the concentration of these agonists could overcome the defect. Platelet aggregation induced by collagen, a non-GPCR agonist, was also compromised in the absence of P-Rex1. Along with these phenotypic changes were impaired Rac1 activation; reduced ATP secretion; and decreased phosphorylation of Akt, c-Jun N-terminal kinase, and p38 mitogen-activated protein kinase in P-Rex1-/- platelets on agonist stimulation. CONCLUSION: These results demonstrate for the first time the presence of P-Rex1 in platelets as well as its role in platelet secretion and aggregation induced by low-dose agonists for GPCR and by collagen.


Assuntos
Plaquetas/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Agregação Plaquetária , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Tempo de Sangramento , Plaquetas/efeitos dos fármacos , Western Blotting , Colágeno/farmacologia , Grânulos Citoplasmáticos/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Feminino , Genótipo , Fatores de Troca do Nucleotídeo Guanina/deficiência , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Hemostasia/genética , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Fosforilação , Agregação Plaquetária/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Trombina/metabolismo , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
5.
Mol Pharmacol ; 80(6): 965-78, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21810922

RESUMO

2-Aminoethyl methylphosphonate (2-AEMP), an analog of GABA, has been found to exhibit antagonist activity at GABA(A)-ρ1 (also known as ρ1 GABA(C)) receptors. The present study was undertaken to elucidate 2-AEMP's action and to test the activities of 2-AEMP analogs. Whole-cell patch-clamp techniques were used to record membrane currents in neuroblastoma cells stably transfected with human GABA(A)-ρ1 receptors. The action of 2-AEMP was compared with that of 1,2,5,6-tetrahydropyridin-4-yl methylphosphinic acid (TPMPA), a commonly used GABA(A)-ρ1 antagonist. With 10 µM GABA, 2-AEMP's IC(50) (18 µM) differed by less than 2.5-fold from that of TPMPA (7 µM), and results obtained were consistent with a primarily competitive mode of inhibition by 2-AEMP. Terminating the presentation of 2-AEMP or TPMPA in the presence of GABA produced a release from inhibition. However, the rate of inhibition release upon the termination of 2-AEMP considerably exceeded that determined with termination of TPMPA. Moreover, when presented at concentrations near their respective IC(50) values, the preincubation period associated with 2-AEMP's onset of inhibition was much shorter than that for TPMPA. Analogs of 2-AEMP possessing a benzyl or n-butyl rather than a methyl substituent at the phosphorus atom, as well as analogs bearing a C-methyl substituent on the aminoethyl side chain, exhibited reduced potency relative to 2-AEMP. Of these analogs, only (R)-2-aminopropyl methylphosphonate significantly diminished the response to 10 µM GABA. Structure-activity relationships are discussed in the context of molecular modeling of ligand binding to the antagonist binding site of the GABA(A)-ρ1 receptor.


Assuntos
Antagonistas de Receptores de GABA-A/química , Antagonistas de Receptores de GABA-A/metabolismo , Compostos Organofosforados/química , Compostos Organofosforados/metabolismo , Receptores de GABA-A/metabolismo , Receptores de GABA/metabolismo , Animais , Sítios de Ligação/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Feminino , Antagonistas de Receptores de GABA-A/farmacologia , Humanos , Compostos Organofosforados/farmacologia , Fatores de Tempo , Xenopus laevis
6.
Exp Eye Res ; 93(1): 59-64, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21536029

RESUMO

The GABA(C) receptor, a postsynaptic membrane receptor expressed prominently in the retina, is a ligand-gated ion channel that consists of a combination of ρ subunits. We report characterization of a novel guinea pig polyclonal antibody, termed GABA(C) Ab N-14, directed against a 14-mer peptide (N-14) of the extracellular domain of the human ρ1 subunit. The antibody exhibits high sensitivity for N-14 by ELISA. In Western blots, GABA(C) Ab N-14 shows reactivity with the ρ1 subunit of preparations obtained from ρ1 GABA(C)-expressing neuroblastoma cells, Xenopus oocytes, and mammalian retina and brain. Flow cytometry reveals a rightward shift in mean fluorescence intensity of GABA(C)-expressing neuroblastoma cells probed with GABA(C) Ab N-14. Immunostaining of neuroblastoma cells and oocytes with GABA(C) Ab N-14 yields fluorescence only with GABA(C)-expressing cells. Antibody binding has no effect on GABA-elicited membrane current responses. Immunostaining of human retinal sections shows preferential staining within the inner plexiform layer. GABA(C) Ab N-14 appears well suited for investigative studies of GABA(C) ρ1 subunit in retina and other neural tissues.


Assuntos
Anticorpos/sangue , Imunoglobulina G/imunologia , Receptores de GABA/imunologia , Retina/imunologia , Sequência de Aminoácidos , Animais , Especificidade de Anticorpos/imunologia , Western Blotting , Encéfalo/imunologia , Cromatografia de Afinidade , Reações Cruzadas/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Técnica Indireta de Fluorescência para Anticorpo , Cobaias , Humanos , Dados de Sequência Molecular , Neuroblastoma/imunologia , Oócitos/imunologia , Fragmentos de Peptídeos/imunologia , Subunidades Proteicas/imunologia , Células Tumorais Cultivadas , Xenopus laevis
7.
Cell Signal ; 18(4): 564-76, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16125366

RESUMO

The present study provides evidence that G protein coupled receptor (GPCR) signaling pathways participate in an interactive signaling network governed by the principles of mass action. Using an inducible thromboxane A2 receptor (TPR)/platelet activating factor receptor (PAFR) co-expressing cell model, TPR or PAFR expression was independently up-regulated. Immunostaining and radioligand binding experiments demonstrated that this receptor up-regulation resulted in increased GPCR:G protein mass ratios. This increase in mass ratio impacted both TPR and PAFR ligand affinity. Specifically, up-regulating TPR expression not only decreased TPR ligand affinity, but also decreased the ligand affinity of PAFRs. A similar effect on ligand affinities was observed when PAFRs were up-regulated. In addition, increasing the GPCR:G protein mass ratio for TPRs led to desensitization of the calcium mobilization response to PAFR activation, and increasing PAFR mass desensitized the TPR-mediated calcium response. Finally, it was observed that an increased TPR:G protein mass ratio was associated with a shift in the TPR signaling response, and revealed an additional TPR signaling pathway through G(S). Collectively, these results describe a novel mechanism, i.e., mass-dependent GPCR signaling, by which cells can modulate their GPCR signaling pathways and signaling priorities.


Assuntos
Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Animais , Azepinas/farmacologia , Plaquetas/efeitos dos fármacos , Células CHO , Cálcio/metabolismo , Cricetinae , Humanos , Ligantes , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores de Tromboxano A2 e Prostaglandina H2/agonistas , Receptores de Tromboxano A2 e Prostaglandina H2/antagonistas & inibidores , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Triazóis/farmacologia , Regulação para Cima
8.
Cell Signal ; 16(5): 521-33, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14751539

RESUMO

Thromboxane A2 receptors (TPs) are widely distributed among different organ systems and have been localized on both cell membranes and intracellular structures. Following the initial cloning of this receptor class from human placenta, the deduced amino acid sequence predicted seven-transmembrane spanning regions, four extracellular domains and four intracellular domains, making TP a member of the seven-transmembrane G-protein-coupled receptor (GPCR) super family. A single gene on chromosome 19p13.3 leads to the expression of two separate TP isoforms: TPalpha which is broadly expressed in numerous tissues, and a splice variant termed TPbeta which may have a more limited tissue distribution. Mutagenesis, photoaffinity labelling, and immunological studies have indicated that the ligand binding domains for this receptor may reside in both the transmembrane (TM) and extracellular regions of the receptor protein. In addition, separate studies have provided evidence that this receptor can couple to at least four separate G protein families. As a consequence, TP signalling has been shown to result in a broad range of cellular responses including phosphoinositide metabolism, calcium redistribution, cytoskeletal arrangement, integrin activation, kinase activation, and the subsequent nuclear signalling events involved in DNA synthesis, cell proliferation, cell survival and cell death. While activation of these different signalling cascades can all derive from TP stimulation, the relative signalling preference for a given cascade appears to be both tissue and cell specific. Finally, separate studies have indicated that TP signalling capacity can be both down-regulated by protein kinase activation and up-regulated by GPCR cross-signalling. Thus, the multitude of signalling events which derive from TP activation can themselves be modulated by endogenous cellular messengers.


Assuntos
Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Transdução de Sinais/fisiologia , Tromboxano A2/metabolismo , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Antagonistas de Prostaglandina/farmacologia
9.
J Biol Chem ; 284(24): 16108-16117, 2009 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-19346255

RESUMO

ADP plays an integral role in the process of hemostasis by signaling through two platelet G-protein-coupled receptors, P2Y1 and P2Y12. The recent use of antagonists against these two receptors has contributed a substantial body of data characterizing the ADP signaling pathways in human platelets. Specifically, the results have indicated that although P2Y1 receptors are involved in the initiation of platelet aggregation, P2Y12 receptor activation appears to account for the bulk of the ADP-mediated effects. Based on this consideration, emphasis has been placed on the development of a new class of P2Y12 antagonists (separate from clopidogrel and ticlopidine) as an approach to the treatment of thromboembolic disorders. The present work examined the molecular mechanisms by which two of these widely used adenosine-based P2Y12 antagonists (2-methylthioadenosine 5'-monophosphate triethylammonium salt (2MeSAMP) and ARC69931MX), inhibit human platelet activation. It was found that both of these compounds raise platelet cAMP to levels that substantially inhibit platelet aggregation. Furthermore, the results demonstrated that this elevation of cAMP did not require Gi signaling or functional P2Y12 receptors but was mediated through activation of a separate G protein-coupled pathway, presumably involving Gs. However, additional experiments revealed that neither 2MeSAMP nor ARC69931MX (cangrelor) increased cAMP through activation of A2a, IP, DP, or EP2 receptors, which are known to couple to Gs. Collectively, these findings indicate that 2MeSAMP and ARC69931MX interact with an unidentified platelet G protein-coupled receptor that stimulates cAMP-mediated inhibition of platelet function. This inhibition is in addition to that derived from antagonism of P2Y12 receptors.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/análogos & derivados , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Agregação Plaquetária/efeitos dos fármacos , Antagonistas do Receptor Purinérgico P2 , Transdução de Sinais/efeitos dos fármacos , Tionucleotídeos/farmacologia , Difosfato de Adenosina/metabolismo , Difosfato de Adenosina/farmacologia , Monofosfato de Adenosina/farmacologia , Trifosfato de Adenosina/farmacologia , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/farmacologia , Humanos , Técnicas In Vitro , Receptor A2A de Adenosina/metabolismo , Receptor PAR-1/metabolismo , Receptores de Epoprostenol , Receptores Imunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Receptores de Prostaglandina E/metabolismo , Receptores de Prostaglandina E Subtipo EP2 , Receptores Purinérgicos P2Y12 , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo
10.
Mol Cell Biol ; 28(20): 6329-41, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18710937

RESUMO

The present study investigated G protein expression, localization, and functional coupling to thromboxane A(2) receptors (TPRs) during oligodendrocyte (OLG) development. It was found that as OLGs mature, the expression levels of G(q) increase while those of G(13) decrease. In contrast, the expression levels of G(s), G(o), and G(i) do not change significantly. Localization studies revealed that G(q), G(13), and G(i) are present only in the extranuclear compartment, whereas G(s) and G(o) are found in both the extranuclear and the nuclear compartments. Purification of TPR-G protein complexes demonstrated that TPRs couple to both G(q) and G(13) in the extranuclear compartment but only to G(s) in the nuclear compartment. Furthermore, functional analysis revealed that stimulation of nuclear TPR in OLGs stimulates CREB phosphorylation and myelin basic protein transcription and increases survival. Collectively, these results demonstrate that (i) OLGs selectively modulate the expression of certain G proteins during development, (ii) G proteins are differentially localized in OLGs leading to subcellular compartmentalization, (iii) TPRs couple to G(q) and G(13) in the extranuclear compartment and to G(s) only in the nucleus, (iv) mature OLGs have a functional nuclear TPR-G(s) signaling pathway, and (v) nuclear TPR signaling can stimulate CREB phosphorylation and myelin gene transcription and increase cell survival. These findings represent a novel paradigm for selective modulation of G protein-coupled receptor-G protein signaling during cell development.


Assuntos
Compartimento Celular , Diferenciação Celular , Núcleo Celular/metabolismo , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Transdução de Sinais , Animais , AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Ligação ao GTP/isolamento & purificação , Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica , Espaço Intracelular/metabolismo , Proteína Básica da Mielina/genética , Proteína Básica da Mielina/metabolismo , Fosforilação , Transporte Proteico , Ratos , Ratos Sprague-Dawley , Receptores de Tromboxano A2 e Prostaglandina H2/genética , Receptores de Tromboxano A2 e Prostaglandina H2/isolamento & purificação , Células-Tronco/citologia , Células-Tronco/metabolismo , Tromboxano A2/biossíntese
11.
Biochem Pharmacol ; 75(12): 2301-15, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18455148

RESUMO

Since isoprostanes are thought to participate in the pathogenesis of thrombosis, presumably through their interaction with thromboxane receptors (TPRs), we examined the ability of 8-iso-PGF(2alpha) to bind/signal through TPRs. Using TPR expressing HEK cells, it was found that 8-iso-PGF(2alpha) mobilized calcium and bound TPRs with a dissociation constant (K(d)) of 57 nM. Interestingly, site-directed-mutagenesis revealed that 8-iso-PGF(2alpha) has a unique coordination profile with TPRs. Thus, while Phe184 and Asp193 are shared by both 8-iso-PGF(2alpha) and classical TPR ligands, Phe196 was found to be required only for 8-iso-PGF(2alpha) binding. Functional studies also revealed interesting results. Namely, that 8-iso-PGF(2alpha) signals in human platelets through both a stimulatory (TPR-dependent) and an inhibitory (cAMP-dependent) pathway. Consistent with the existence of two signaling pathways, platelets were also found to possess two separate binding sites for 8-iso-PGF(2alpha). While the stimulatory site is represented by TPRs, the second cAMP inhibitory site is presently unidentified, but does not involve receptors for PGI(2), PGD(2) or PGE(2). In summary, these studies provide the first documentation that: (1) 8-iso-PGF(2alpha) coordinates with Phe184, Asp193 and Phe196 on platelet TPRs; (2) Phe196 serves as a unique TPR binding site for 8-iso-PGF(2alpha); (3) 8-iso-PGF(2alpha) signals through both stimulatory and inhibitory pathways in platelets; (4) 8-iso-PGF(2alpha) inhibits human platelet activation through a cAMP-dependent mechanism; (5) 8-iso-PGF(2alpha) interacts with platelets at two separate binding sites. Collectively, these results provide evidence for a novel isoprostane function in platelets which is mediated through a cAMP-coupled receptor.


Assuntos
Plaquetas/metabolismo , AMP Cíclico/metabolismo , Dinoprosta/análogos & derivados , Isoprostanos/metabolismo , Ativação Plaquetária , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Sítios de Ligação , Plaquetas/citologia , Plaquetas/fisiologia , Cálcio/metabolismo , Linhagem Celular , Forma Celular/fisiologia , Citosol/metabolismo , Dinoprosta/metabolismo , Humanos , Ligação de Hidrogênio , Ligantes , Mutagênese Sítio-Dirigida , Ativação Plaquetária/fisiologia , Agregação Plaquetária/fisiologia , Ensaio Radioligante , Receptores de Tromboxano A2 e Prostaglandina H2/genética , Transdução de Sinais , Transfecção
12.
J Biol Chem ; 282(14): 10210-22, 2007 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-17298951

RESUMO

This study investigated the involvement of Galpha(13) switch region I (SRI) in protease-activated receptor 1 (PAR1)-mediated platelet function and signaling. To this end, myristoylated peptides representing the Galpha(13) SRI (Myr-G(13)SRI(pep)) and its random counterpart were evaluated for their effects on PAR1 activation. Initial studies demonstrated that Myr-G(13)SRI(pep) and Myr-G(13)SRI(Random-pep) were equally taken up by human platelets and did not interfere with PAR1-ligand interaction. Subsequent experiments revealed that Myr-G(13)SRI(pep) specifically bound to platelet RhoA guanine nucleotide exchange factor (p115RhoGEF) and blocked PAR1-mediated RhoA activation in platelets and human embryonic kidney cells. These results suggest a direct interaction of Galpha(13) SRI with p115RhoGEF and a mechanism for Myr-G(13)SRI(pep) inhibition of RhoA activation. Platelet function studies demonstrated that Myr-G(13)SRI(pep) specifically inhibited PAR1-stimulated shape change, aggregation, and secretion in a dose-dependent manner but did not inhibit platelet activation induced by either ADP or A23187. It was also found that Myr-G(13)SRI(pep) inhibited low dose, but not high dose, thrombin-induced aggregation. Additional experiments showed that PAR1-mediated calcium mobilization was partially blocked by Myr-G(13)SRI(pep) but not by the Rho kinase inhibitor Y-27632. Finally, Myr-G(13)SRI(pep) effectively inhibited PAR1-induced stress fiber formation and cell contraction in endothelial cells. Collectively, these results suggest the following: 1) interaction of Galpha(13) SRI with p115RhoGEF is required for G(13)-mediated RhoA activation in platelets; 2) signaling through the G(13) pathway is critical for PAR1-mediated human platelet functional changes and low dose thrombin-induced aggregation; and 3) G(13) signaling elicits calcium mobilization in human platelets through a Rho kinase-independent mechanism.


Assuntos
Plaquetas/metabolismo , Sinalização do Cálcio/fisiologia , Forma Celular/fisiologia , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Peptídeos/metabolismo , Agregação Plaquetária/fisiologia , Difosfato de Adenosina/farmacologia , Plaquetas/citologia , Calcimicina/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/farmacologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Hemostáticos/farmacologia , Humanos , Ionóforos/farmacologia , Rim/citologia , Rim/metabolismo , Peptídeos/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Receptor PAR-1 , Fatores de Troca de Nucleotídeo Guanina Rho , Trombina/farmacologia , Proteína rhoA de Ligação ao GTP/metabolismo
13.
J Biol Chem ; 281(37): 26951-65, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16837469

RESUMO

Despite the well documented involvement of thromboxane A(2) receptor (TPR) signaling in the pathogenesis of thrombotic diseases, there are currently no rationally designed antagonists available for clinical use. To a large extent, this derives from a lack of knowledge regarding the topography of the TPR ligand binding pocket. On this basis, the purpose of the current study was to identify the specific amino acid residues in the TPR protein that regulate ligand coordination and binding. The sites selected for mutation reside within or in close proximity to a region we previously defined as a TPR ligand binding region (i.e. the C terminus of the second extracellular loop and the leading edge of the fifth transmembrane domain). Mutation of these residues caused varying effects on the TPR-ligand coordination process. Specifically, the D193A, D193Q, and D193R mutants lost SQ29,548 (antagonist) binding and exhibited a dramatically reduced calcium response, which could not be restored by elevated U46619 (agonist) doses. The F184Y mutant lost SQ29,548 binding and exhibited a reduced calcium response (which could be restored by elevated U46619); and the T186A and S191T mutants lost SQ29,548 binding and retained a normal U46619-induced calcium response. Furthermore, these last three mutants also revealed a divergence in the binding of two structurally different antagonists, SQ29,548 and BM13.505. Two separate mutants that exhibited SQ29,548 binding yielded either a normal (F196Y) or reduced (S201T) U46619 response. Finally, mutation of other residues directly adjacent to those described above (e.g. E190A and F200A) produced no detectable effects on either SQ29,548 binding or the U46619-induced response. In summary, these results identify key amino acids (in particular Asp(193)) involved in TPR ligand coordination. These findings also demonstrate that TPR-specific ligands interact with different residues in the ligand-binding pocket.


Assuntos
Receptores de Tromboxano A2 e Prostaglandina H2/química , Sequência de Aminoácidos , Aminoácidos/química , Ácido Aspártico/química , Cálcio/química , Linhagem Celular , Relação Dose-Resposta a Droga , Humanos , Ligantes , Dados de Sequência Molecular , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Receptores de Tromboxano A2 e Prostaglandina H2/agonistas , Receptores de Tromboxano A2 e Prostaglandina H2/antagonistas & inibidores
14.
J Neurosci Res ; 84(7): 1402-14, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16998891

RESUMO

The present work investigates the role of thromboxane A(2) (TXA(2)) receptors in the development of oligodendrocytes (OLGs). The results demonstrate that the proteins of the TXA(2) signaling pathway, i.e., cyclooxygenase (COX-1), TXA(2) synthase (TS), and TXA(2) receptor (TPR) are expressed in the developing rat brain during myelination. Furthermore, culture of OLG progenitor cells (OPCs) revealed that the expression levels of these proteins as well as TXA(2) synthesis increase during OLG maturation. Separate studies established that activation of TPRs by the agonist U46619 increases intracellular calcium in both OPCs and OLGs as visualized by digital fluorescence imaging. Immunocytochemical staining demonstrated that TPRs are localized in the plasma membrane and perinuclear compartments in OPCs. However, during OLG differentiation, TPRs shift their localization pattern and also become associated with the nuclear compartment. This shift to nuclear localization was confirmed by biochemical analysis in cultured cells and by immunocytochemical analysis in developing rat brain. Finally, it was found that U46619 activation of TPRs in maturing OLGs resulted in enhanced myelin basic protein (MBP) expression. Alternatively, inhibition of endogenous TPR signaling led to reduced MBP expression. Furthermore, TPR-mediated MBP expression was found to be associated with increased transcription from the MBP promoter using a MBP-luciferase reporter. Collectively, these findings suggest a novel TPR signaling pathway in OLGs and a potential role for this signaling during OLG maturation and myelin production.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteína Básica da Mielina/metabolismo , Oligodendroglia/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Tromboxano A2 e Prostaglandina H2/fisiologia , Transdução de Sinais/fisiologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Fatores Etários , Animais , Animais Recém-Nascidos , Western Blotting/métodos , Encéfalo/citologia , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , AMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , Gangliosídeos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/fisiologia , Tromboxano B2/metabolismo , Fatores de Tempo , Transfecção/métodos
15.
J Neurochem ; 93(2): 257-68, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15816849

RESUMO

Thromboxane A(2) receptors (TP) were previously localized to discrete regions in the rat brain on myelinated fiber tracts and oligodendrocytes (OLGs). The present studies extended these findings and investigated the effects of TP signaling on cell proliferation, survival, and gene expression in OLG progenitor cells (OPCs) and OLGs. It was found that the TP agonist, U46619 stimulated the proliferation of OPCs and promoted the survival of mature OLGs. Examination of the early gene expression events involved in OPC proliferation, revealed that c-fos expression was substantially increased by U46619 stimulation. Treatment of OPCs or OLGs with U46619 caused activation of the mitogen-activated protein kinases (MAPK) ERK 1/2. In OPCs this activation was blocked by inhibition of src. However, in OLGs this phosphorylation was not only blocked by inhibition of src but also by inhibition of protein kinase C (PKC). Furthermore, U46619 was found to increase CREB phosphorylation in both OPCs and OLGs. Similar to ERK 1/2 activation, there was a divergence in the mechanism of the TP-mediated CREB response for each cell type. Specifically, U46619 activation was attenuated by src and protein kinase A (PKA) inhibition in OPCs, whereas in OLGs this effect was blocked by inhibition of src, PKA as well as by inhibition of PKC. Collectively, these results provide the first demonstration that TP-activated nuclear signaling events are involved in the proliferation of OPCs, the survival of mature OLGs, and the stimulation of gene expression.


Assuntos
Proliferação de Células/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Receptores de Tromboxano A2 e Prostaglandina H2/genética , Receptores de Tromboxano A2 e Prostaglandina H2/fisiologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Tromboxano A2 e Prostaglandina H2/agonistas , Receptores de Tromboxano A2 e Prostaglandina H2/biossíntese
16.
J Biol Chem ; 277(19): 16791-7, 2002 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-11877412

RESUMO

The human thromboxane A(2) (TP) receptor, a member of the G protein-coupled receptor superfamily, consists of seven transmembrane segments. Attempts to elucidate the specific segment(s) that define the receptor ligand-binding pocket have produced less than definitive and sometimes conflicting results. On this basis, the present work identified an amino acid sequence of the TP receptor that is directly involved in ligand binding. Mapping of this domain was confirmed by two separate approaches: photoaffinity labeling and site-specific antibodies. The newly synthesized, biotinylated photoaffinity probe, SQBAzide, was first shown to specifically label TP receptor protein. Sequential digestion of this protein with CNBr/trypsin revealed photolabeling of a 2.9-kDa peptide. Using anti-peptide antibodies directed against different regions of the receptor protein, it was established that this peptide represents the predicted cleavage product for CNBr/trypsin and corresponds to amino acids Arg(174)-Met(202) of the receptor protein. Furthermore, antibody screening revealed that inhibition of the amino acid region Cys(183)-Asp(193) was critical for radioligand binding and platelet aggregation, whereas inhibition of Gly(172)-Cys(183) was not. Collectively these findings provide evidence that ligands interact with amino acids contained within the C-terminal portion of the third extracellular domain (ED3) of the receptor protein. This information should be of significant value in the study of TP receptor structure and signaling.


Assuntos
Receptores de Tromboxanos/química , Sequência de Aminoácidos , Ácido Aspártico/química , Sítios de Ligação , Ligação Competitiva , Plaquetas/metabolismo , Cisteína/química , Relação Dose-Resposta Imunológica , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Glicina/química , Humanos , Ligantes , Modelos Químicos , Dados de Sequência Molecular , Agregação Plaquetária , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Receptores de Tromboxanos/metabolismo , Transdução de Sinais , Espectrometria de Fluorescência , Tripsina/química , Tripsina/metabolismo , Tripsina/farmacologia
17.
J Biol Chem ; 278(1): 124-30, 2003 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-12399457

RESUMO

The present studies mapped the protein kinase A (PKA) phosphorylation site of Galpha(13) and studied the consequences of its phosphorylation. Initial experiments using purified human Galpha(13) and the PKA catalytic subunit established that PKA directly phosphorylates Galpha(13). The location of this phosphorylation site was next investigated with a new synthetic peptide (G(13)SRI(pep)) containing the PKA consensus sequence (Arg-Arg-Pro-Thr(203)) within the switch I region of Galpha(13). G(13)SRI(pep) produced a dose-dependent inhibition of PKA-mediated Galpha(13) phosphorylation. On the other hand, the Thr-phosphorylated derivative of G(13)SRI(pep) possessed no inhibitory activity, suggesting that Galpha(13) Thr(203) may represent the phosphorylation site. Confirmation of this notion was obtained by showing that the Galpha(13)-T203A mutant (in COS-7 cells) could not be phosphorylated by PKA. Additional studies using co-elution affinity chromatography and co-immunoprecipitation demonstrated that Galpha(13) phosphorylation stabilized coupling of Galpha(13) with platelet thromboxane A(2) receptors but destabilized coupling of Galpha(13) to its betagamma subunits. In order to determine the functional consequences of this phosphorylation on Galpha(13) signaling, activation of the Rho pathway was investigated. Specifically, Chinese hamster ovary cells overexpressing human Galpha(13) wild type (Galpha(13)-WT) or Galpha(13)-T203A mutant were generated and assayed for Rho activation. It was found that 8-bromo-cyclic AMP caused a significant decrease (50%; p < 0.002) of Rho activation in Galpha(13) wild type cells but produced no change of basal Rho activation levels in the mutant (p > 0.4). These results therefore suggest that PKA blocks Rho activation by phosphorylation of Galpha(13) Thr(203).


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Plaquetas/metabolismo , Bovinos , Linhagem Celular , Sequência Consenso , AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/genética , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP , Humanos , Substâncias Macromoleculares , Mutagênese Sítio-Dirigida , Fosforilação , Ligação Proteica , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Receptores de Tromboxanos/metabolismo , Sistemas do Segundo Mensageiro/fisiologia
18.
J Biol Chem ; 278(33): 30725-31, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12796499

RESUMO

Thromboxane A2 (TXA2)-mediated platelet secretion and aggregation are important in thrombosis. Here, we present a novel finding that the stable TXA2 analogue, U46619, induces two waves of platelet secretion, each of which precedes a distinct wave of platelet aggregation. ADP released from platelets during the first wave of secretion played a major role in augmenting the first wave of platelet aggregation. The second wave of platelet secretion and aggregation required the first wave of both ADP secretion and aggregation and were blocked by either the integrin inhibitor RGDS or a P2Y12 receptor antagonist, indicating a requirement for both the integrin outside-in signal and ADP-activated Gi pathway. U46619 stimulated phosphoinositide 3-kinase (PI3K)-dependent phosphorylation of Akt, which was augmented by ADP but did not require integrin outside-in signaling. Platelets from PI3Kgamma knock-out mice or PI3K inhibitor-treated platelets showed an impaired second wave of platelet secretion and aggregation. However, the second wave of platelet aggregation was restored by addition of exogenous ADP to PI3Kgamma deficient or PI3K inhibitor-treated platelets. Thus, our data indicate that PI3K, together with the integrin outside-in signaling, play a central role in inducing the second wave of platelet secretion, which leads to the second wave of irreversible platelet aggregation.


Assuntos
Plaquetas/enzimologia , Plaquetas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Receptores de Tromboxanos/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Difosfato de Adenosina/metabolismo , Difosfato de Adenosina/farmacologia , Animais , Plaquetas/efeitos dos fármacos , Humanos , Integrinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfatidilinositol 3-Quinases/genética , Fosforilação/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Vasoconstritores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA