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










Base de dados
Intervalo de ano de publicação
1.
FASEB J ; 29(3): 1056-68, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25466898

RESUMO

Multiple human malignancies rely on C-X-C motif chemokine receptor type 4 (CXCR4) and its ligand, SDF-1/CXCL12 (stroma cell-derived factor 1/C-X-C motif chemokine 12), to metastasize. CXCR4 inhibitors promote the mobilization of bone marrow stem cells, limiting their clinical application for metastasis prevention. We investigated the CXCR4-initiated signaling circuitry to identify new potential therapeutic targets. We used HeLa human cancer cells expressing high levels of CXCR4 endogenously. We found that CXCL12 promotes their migration in Boyden chamber assays and single cell tracking. CXCL12 activated mTOR (mechanistic target of rapamycin) potently in a pertussis-sensitive fashion. Inhibition of mTOR complex 1 (mTORC1) by rapamycin [drug concentration causing 50% inhibition (IC50) = 5 nM] and mTORC1/mTORC2 by Torin2 (IC50 = 6 nM), or by knocking down key mTORC1/2 components, Raptor and Rictor, respectively, decreased directional cell migration toward CXCL12. We developed a CXCR4-mediated spontaneous metastasis model by implanting HeLa cells in the tongue of SCID-NOD mice, in which 80% of the animals develop lymph node metastasis. It is surprising that mTORC1 disruption by Raptor knockdown was sufficient to reduce tumor growth by 60% and spontaneous metastasis by 72%, which were nearly abolished by rapamycin. In contrast, disrupting mTORC2 had no effect in tumor growth or metastasis compared with control short hairpin RNAs. These data suggest that mTORC1 may represent a suitable therapeutic target in human malignancies using CXCR4 for their metastatic spread. .


Assuntos
Movimento Celular , Quimiocina CXCL12/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Complexos Multiproteicos/metabolismo , Receptores CXCR4/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Neoplasias do Colo do Útero/secundário , Animais , Apoptose , Western Blotting , Proliferação de Células , Feminino , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Metástase Linfática , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transdução de Sinais , Células Tumorais Cultivadas , Neoplasias do Colo do Útero/metabolismo
2.
Mol Cell ; 49(1): 94-108, 2013 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-23177739

RESUMO

Activating mutations in GNAQ and GNA11, encoding members of the Gα(q) family of G protein α subunits, are the driver oncogenes in uveal melanoma, and mutations in Gq-linked G protein-coupled receptors have been identified recently in numerous human malignancies. How Gα(q) and its coupled receptors transduce mitogenic signals is still unclear because of the complexity of signaling events perturbed upon Gq activation. Using a synthetic-biology approach and a genome-wide RNAi screen, we found that a highly conserved guanine nucleotide exchange factor, Trio, is essential for activating Rho- and Rac-regulated signaling pathways acting on JNK and p38, and thereby transducing proliferative signals from Gα(q) to the nucleus independently of phospholipase C-ß. Indeed, whereas many biological responses elicited by Gq depend on the transient activation of second-messenger systems, Gq utilizes a hard-wired protein-protein-interaction-based signaling circuitry to achieve the sustained stimulation of proliferative pathways, thereby controlling normal and aberrant cell growth.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/fisiologia , Mitose , Proteínas Serina-Treonina Quinases/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Fator de Transcrição AP-1/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Clozapina/análogos & derivados , Clozapina/farmacologia , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Ativação Enzimática , Feminino , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Técnicas de Silenciamento de Genes , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Camundongos , Camundongos Nus , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mitógenos/farmacologia , Células NIH 3T3 , Transplante de Neoplasias , Neoplasias/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , Receptores Acoplados a Proteínas G/genética
3.
Cancer Res ; 71(22): 7103-12, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21975930

RESUMO

Despite our improved understanding of cancer, the 5-year survival rate for head and neck squamous cell carcinomas (HNSCC) patients remains relatively unchanged at 50% for the past three decades. HNSCCs often metastasize to locoregional lymph nodes, and lymph node involvement represents one of the most important prognostic factors of poor clinical outcome. Among the multiple dysregulated molecular mechanism in HNSCCs, emerging basic, preclinical, and clinical findings support the importance of the mTOR signaling route in HNSCC progression. Indeed, we observed here that the activation of mTOR is a widespread event in clinical specimens of HNSCCs invading locoregional lymph nodes. We developed an orthotopic model of HNSCC consisting of the implantation of HNSCC cells into the tongues of immunocompromised mice. These orthotopic tumors spontaneously metastasize to the cervical lymph nodes, where the presence of HNSCC cells can be revealed by histologic and immunohistochemical evaluation. Both primary and metastatic experimental HNSCC lesions exhibited elevated mTOR activity. The ability to monitor and quantitate lymph node invasion in this model system enabled us to explore whether the blockade of mTOR could impact HNSCC metastasis. We found that inhibition of mTOR with rapamycin and the rapalog RAD001 diminished lymphangiogenesis in the primary tumors and prevented the dissemination of HNSCC cancer cells to the cervical lymph nodes, thereby prolonging animal survival. These findings may provide a rationale for the future clinical evaluation of mTOR inhibitors, including rapamycin and its analogues, as part of a molecular-targeted metastasis preventive strategy for the treatment of patients with HNSCC.


Assuntos
Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Linfangiogênese/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Carcinoma de Células Escamosas/patologia , Everolimo , Feminino , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Metástase Linfática , Camundongos , Camundongos SCID , Sirolimo/análogos & derivados , Sirolimo/uso terapêutico , Carcinoma de Células Escamosas de Cabeça e Pescoço , Serina-Treonina Quinases TOR/fisiologia , Fatores de Transcrição/fisiologia
4.
PLoS One ; 6(2): e16586, 2011 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-21347357

RESUMO

BACKGROUND: We have previously shown that ADP-induced TXA(2) generation requires signaling from αIIbß3 integrin in platelets. Here we observed that, unlike ADP, protease-activated receptor (PAR)-mediated TXA(2) generation occurs independently of αIIbß3. PAR agonists, but not ADP, activate G(12/13) signaling pathways. Hence, we evaluated the role of these pathways in TXA(2) generation. PRINCIPAL FINDINGS: Inhibition of ADP-induced thromboxane generation by fibrinogen receptor antagonist SC57101 was rescued by co-stimulation of G(12/13) pathways with YFLLRNP. This observation suggested an existence of a common signaling effector downstream of integrins and G(12/13) pathways. Hence, we evaluated role of three potential tyrosine kinases; c-Src, Syk and FAK (Focal Adhesion Kinase) that are known to be activated by integrins. c-Src and Syk kinase did not play a role in ADP-induced functional responses in platelets. Selective activation of G(12/13) pathways resulted in the activation of FAK, in the absence of integrin signaling. Interestingly, αIIbß3-mediated FAK activation occurred in a Src family kinase (SFK)-independent manner whereas G(12/13) pathway caused FAK activation in a SFK and RhoA-dependent manner. A FAK selective inhibitor TAE-226, blocked TXA(2) generation. However, in comparison to WT mice, Pf4-Cre/Fak-Floxed mice did not show any difference in platelet TXA(2) generation. CONCLUSIONS: Therefore, we conclude that differential activation of FAK occurs downstream of Integrins and G(12/13) pathways. However, the common effector molecule, possibly a tyrosine kinase downstream of integrins and G(12/13) pathways contributing to TXA(2) generation in platelets remains elusive.


Assuntos
Plaquetas/citologia , Plaquetas/metabolismo , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Transdução de Sinais , Tromboxano A2/biossíntese , Difosfato de Adenosina/metabolismo , Animais , Plaquetas/enzimologia , Ativação Enzimática , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Quinase Syk , Quinases Associadas a rho/metabolismo
5.
Platelets ; 19(8): 614-23, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19012179

RESUMO

Thrombin-induced platelet activation leads to tyrosine phosphorylation of hematopoietic lineage cell-specific protein-1 (HS1), a 75 kDa adapter protein expressed exclusively in cells of hematopoietic lineage. We have shown HS1 to be a functionally important signaling molecule downstream of PAR-4 and GPVI collagen receptor. We have thus begun to elucidate PAR signaling pathway of HS1 phosphorylation, and its functional implications. PAR-1 and PAR-4 activating peptides (SFLLRN and AYPGKF, respectively) induced HS1 phosphorylation in a Gq-dependent manner as shown by incubation with the Gq inhibitor, YM254890. Consistently, HS1 phosphorylation was abolished in platelets from Gq deficient mice upon AYPGKF stimulation. Treatment with ADP receptor antagonists did not affect HS1 phosphorylation. Pretreatment of platelets with Src kinase inhibitors abolished HS1 phosphorylation. Further Syk activation, as measured by tyrosine phosphorylation of Syk (residues 525/526), in response to PAR activation was abolished in the presence of Src inhibitors. HS1 null mice show inhibition of PAR-mediated thromboxane A2 generation compared to wild type littermates. Phosphorylation of Erk, a key signaling molecule in thromboxane generation, was also diminished in HS1 null mice platelets. Based on these findings, we conclude that tyrosine phosphorylation of HS1 occurs downstream of both PAR-1 and PAR-4. HS1 phosphorylation is a Gq mediated response regulated by Src kinases. Thus, HS1 may mediate PAR-induced thromboxane generation through regulation of Erk phosphorylation.


Assuntos
Plaquetas/metabolismo , Proteínas Sanguíneas/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator Estimulador de Colônias de Granulócitos/fisiologia , Receptor PAR-1/metabolismo , Receptores Ativados por Proteinase/metabolismo , Tromboxano A2/biossíntese , Proteínas Adaptadoras de Transdução de Sinal , Animais , Plaquetas/citologia , Proteínas Sanguíneas/metabolismo , Células Cultivadas , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Fator Estimulador de Colônias de Granulócitos/deficiência , Fator Estimulador de Colônias de Granulócitos/metabolismo , Humanos , Camundongos , Camundongos Knockout , Fosforilação , Transdução de Sinais , Quinases da Família src/metabolismo
6.
Front Biosci ; 13: 433-9, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17981558

RESUMO

Microparticles are shed from the platelet membrane upon platelet activation by strong agonists, and they aid in clot formation. As the P2Y1 and the P2Y12 receptors differentially contribute to different platelet functions, we studied the relative contribution of the P2Y1 and P2Y12 receptors to microparticle formation from platelets. The P2Y12 receptor antagonist AR-C 69931MX, but not the P2Y1 receptor antagonist MRS2179, caused a significant decrease in the number microparticles formed by convulxin and thrombin. In addition, there was no significant decrease in microparticle formation in P2Y1 knockout mouse blood when compared to the wild type mice. These results illustrate that the P2Y12 receptor contributes to microparticle formation from activated platelets by a strong agonist, without any significant involvement of the P2Y1 receptor. We also conclude that there is no correlation in the number of microparticles circulating in vivo between the P2Y1 receptor null mice and the wild type mice under unstimulated conditions. Finally, we conclude that the increased bleeding time in the P2Y1 null mice is due to overall platelet dysfunction and not due to the decrease of circulating microparticles.


Assuntos
Plaquetas/metabolismo , Antagonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2/fisiologia , Animais , Tempo de Sangramento , Citometria de Fluxo , Humanos , Lipídeos/química , Camundongos , Microesferas , Modelos Biológicos , Ativação Plaquetária , Agregação Plaquetária , Agonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2/química , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y1 , Receptores Purinérgicos P2Y12
7.
Blood ; 110(7): 2449-56, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17579181

RESUMO

Collagen activates platelets through an intracellular signaling cascade downstream of glycoprotein VI (GPVI). We have investigated the contribution of hematopoietic lineage cell-specific protein 1 (HS1) downstream of GPVI in platelet activation. Stimulation of GPVI leads to tyrosine phosphorylation of HS1, which is blocked by Src-family kinase inhibitors. Coimmunoprecipitation experiments revealed that HS1 associates with Syk and phosphatidylinositol 3-kinases. HS1-null mice displayed increased bleeding times and increased time to occlusion in the FeCl(3) in vivo thrombosis model compared with their wild-type littermates. In addition, aggregation and secretion responses were diminished in HS1-null mouse platelets after stimulation of GPVI and protease-activated receptor 4 (PAR-4) agonists compared with wild-type littermate mouse platelets. Finally, Akt phosphorylation was diminished after GPVI or PAR-4 stimulation in platelets from HS1-null mice compared with their wild-type littermates. These results demonstrate that phosphorylation of the HS1 protein occurs downstream of GPVI stimulation and that HS1 plays a significant functional role in platelet activation downstream of GPVI and PARs.


Assuntos
Plaquetas/metabolismo , Fator Estimulador de Colônias de Granulócitos/metabolismo , Ativação Plaquetária , Transdução de Sinais , Animais , Fator Estimulador de Colônias de Granulócitos/deficiência , Fator Estimulador de Colônias de Granulócitos/genética , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Ativação Plaquetária/efeitos dos fármacos , Glicoproteínas da Membrana de Plaquetas/metabolismo , Ligação Proteica , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/agonistas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Antagonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2/metabolismo
8.
Nat Rev Cancer ; 7(2): 79-94, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17251915

RESUMO

G-protein-coupled receptors (GPCRs), the largest family of cell-surface molecules involved in signal transmission, have recently emerged as crucial players in tumour growth and metastasis. Malignant cells often hijack the normal physiological functions of GPCRs to survive, proliferate autonomously, evade the immune system, increase their blood supply, invade their surrounding tissues and disseminate to other organs. This Review will address our current understanding of the many roles of GPCRs and their signalling circuitry in tumour progression and metastasis. We will also discuss how interfering with GPCRs might provide unique opportunities for cancer prevention and treatment.


Assuntos
Neoplasias/fisiopatologia , Receptores Acoplados a Proteínas G/fisiologia , Dano ao DNA , Humanos , Inflamação/fisiopatologia , Metástase Neoplásica , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Neovascularização Patológica , Conformação Proteica , Receptores Acoplados a Proteínas G/química , Raios Ultravioleta
9.
Blood ; 106(10): 3410-4, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16020504

RESUMO

The binding of von Willebrand factor (VWF) to the platelet membrane glycoprotein Ib-IX (GPIb-IX) results in platelet activation. In this study, we sought to clarify previous conflicting reports and to elucidate the mechanism of activation and the precise role of extracellular signal-regulated kinase (Erk) in VWF-induced platelet activation. Erk2 is activated in platelets on stimulation with VWF/ristocetin in a time-dependent manner. VWF-induced Erk2 phosphorylation and thromboxane A2 (TXA2) release were completely blocked by PP2, an Src family kinase inhibitor, suggesting that Erk is downstream of Src family kinases. U73122, a phospholipase C inhibitor, also abolished TXA2 generation and Erk phosphorylation. Although VWF fostered the agglutination of platelets regardless of any additional treatment, the inhibition of mitogen-activated protein kinase kinase (MEK) with U0126 abolished VWF-induced platelet aggregation and thromboxane production in non-aspirin-treated washed platelets. However, in platelets treated with aspirin, VWF failed to cause any aggregation. Thus, we conclude that VWF stimulation of platelets results in phospholipase A2 activation through Erk stimulation and that Src family kinases and phospholipase C play essential roles in this event. We further conclude that VWF-induced platelet aggregation does not directly depend on Erk activation but has an absolute requirement for Src/Erk-mediated TXA2 generation.


Assuntos
Sistema de Sinalização das MAP Quinases/fisiologia , Agregação Plaquetária/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Receptores de Fibrinogênio/metabolismo , Tromboxano A2/metabolismo , Fator de von Willebrand/metabolismo , Plaquetas/metabolismo , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Fosforilação/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
10.
Blood ; 106(2): 550-7, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15811957

RESUMO

Thrombin has been known to cause tyrosine phosphorylation of protein kinase C delta (PKCdelta) in platelets, but the molecular mechanisms and function of this tyrosine phosphorylation is not known. In this study, we investigated the signaling pathways used by protease-activated receptors (PARs) to cause tyrosine phosphorylation of PKCdelta and the role of this event in platelet function. PKCdelta was tyrosine phosphorylated by either PAR1 or PAR4 in a concentration- and time-dependent manner in human platelets. In particular, the tyrosine 311 residue was phosphorylated downstream of PAR receptors. Also the tyrosine phosphorylation of PKCdelta did not occur in Galpha(q)-deficient mouse platelets and was inhibited in the presence of a phospholipase C (PLC) inhibitor U73122 and calcium chelator BAPTA (5,5'-dimethyl-bis(o-aminophenoxy)ethane-N, N, N ', N '-tetraacetic acid), suggesting a role for Galpha(q) pathways and calcium in this event. Both PAR1 and PAR4 caused a time-dependent activation of Src (pp60c-src) tyrosine kinase and Src tyrosine kinase inhibitors completely blocked the tyrosine phosphorylation of PKCdelta. Inhibition of tyrosine phosphorylation or the kinase activity of PKCdelta dramatically blocked PAR-mediated thromboxane A2 generation. We conclude that thrombin causes tyrosine phosphorylation of PKCdelta in a calcium- and Src-family kinase-dependent manner in platelets, with functional implications in thromboxane A2 generation.


Assuntos
Plaquetas/metabolismo , Ácido Egtázico/análogos & derivados , Proteína Quinase C/sangue , Trombina/metabolismo , Difosfato de Adenosina/farmacologia , Animais , Plaquetas/efeitos dos fármacos , Quelantes/farmacologia , Ácido Egtázico/farmacologia , Inibidores Enzimáticos/farmacologia , Estrenos/farmacologia , Humanos , Técnicas In Vitro , Camundongos , Modelos Biológicos , Fosforilação , Proteína Quinase C/química , Proteína Quinase C-delta , Pirrolidinonas/farmacologia , Receptor PAR-1/sangue , Receptores de Trombina/sangue , Transdução de Sinais , Treonina/química , Fosfolipases Tipo C/antagonistas & inibidores , Tirosina/química , Quinases da Família src/antagonistas & inibidores
11.
Blood ; 105(7): 2749-56, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15546949

RESUMO

G(12/13) or G(q) signaling pathways activate platelet GPIIb/IIIa when combined with G(i) signaling. We tested whether combined G(i) and G(z) pathways also cause GPIIb/IIIa activation and compared the signaling requirements of these events. Platelet aggregation occurred by combined stimulation of G(i) and G(z) pathways in human platelets and in P2Y1-deficient and G alpha(q)-deficient mouse platelets, confirming that the combination of G(i) and G(z) signaling causes platelet aggregation. When G(i) stimulation was combined with G(z) stimulation, there was a small mobilization of intracellular calcium. Chelation of intracellular calcium decreased the extent of this platelet aggregation, whereas it abolished the G(q) plus G(i)-mediated platelet aggregation. Costimulation of G(i) plus G(z) pathways also caused thromboxane generation that was dependent on outside-in signaling and was inhibited by PP2, a Src family tyrosine kinase inhibitor. Src family tyrosine kinase inhibitors also inhibited platelet aggregation and decreased the PAC-1 binding caused by costimulation of G(i) and G(z) signaling pathways in aspirin-treated platelets. However, Src family kinase inhibitors did not affect G(q) plus G(i)-mediated platelet aggregation. We conclude that the combination of G(i) plus G(z) pathways have different requirements than G(q) plus G(i) pathways for calcium and Src family kinases in GPIIb/IIIa activation and thromboxane production.


Assuntos
Plaquetas/metabolismo , Cálcio/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Tromboxanos/metabolismo , Quinases da Família src/metabolismo , Animais , Aspirina/farmacologia , Plaquetas/efeitos dos fármacos , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/fisiologia , Inibidores da Agregação Plaquetária/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Estimulação Química
12.
J Clin Invest ; 113(3): 340-5, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14755328

RESUMO

Platelet activation occurs in response to vessel injury and is important for the arrest of bleeding. Platelet activation during disease states leads to vascular occlusion and ischemic damage. The P2Y(12) receptor, activated by ADP, plays a central role in platelet activation and is the target of P2Y(12) receptor antagonists that have proven therapeutic value.


Assuntos
Plaquetas/metabolismo , Proteínas de Membrana/metabolismo , Ativação Plaquetária/fisiologia , Receptores Purinérgicos P2/metabolismo , Difosfato de Adenosina/metabolismo , Tempo de Sangramento , Coagulação Sanguínea/fisiologia , Humanos , Receptores Purinérgicos P2Y12
13.
J Biol Chem ; 279(4): 2360-7, 2004 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-14578358

RESUMO

Several platelet agonists, including thrombin, collagen, and thromboxane A(2), cause dense granule release independently of thromboxane generation. Because protein kinase C (PKC) isoforms are implicated in platelet secretion, we investigated the role of individual PKC isoforms in platelet dense granule release. PKCdelta was phosphorylated in a time-dependent manner that coincided with dense granule release in response to protease-activated receptor-activating peptides SFLLRN and AYPGKF in human platelets. Only agonists that caused platelet dense granule secretion activated PKCdelta. SFLLRN- or AYPGKF-induced dense granule release and PKCdelta phosphorylation occurred at the same respective agonist concentration. Furthermore, AYPGKF and SFLLRN-induced dense granule release was blocked by rottlerin, a PKCdelta selective inhibitor. In contrast, convulxin-induced dense granule secretion was potentiated by rottlerin but was abolished by Go6976, a classical PKC isoform inhibitor. However, SFLLRN-induced dense granule release was unaffected in the presence of Go6976. Finally, rottlerin did not affect SFLLRN-induced platelet aggregation, even in the presence of dimethyl-BAPTA, indicating that PKCdelta has no role in platelet fibrinogen receptor activation. We conclude that PKCdelta and the classical PKC isoforms play a differential role in platelet dense granule release mediated by protease-activated receptors and glycoprotein VI. Furthermore, PKCdelta plays a positive role in protease-activated receptor-mediated dense granule secretion, whereas it functions as a negative regulator downstream of glycoprotein VI signaling.


Assuntos
Plaquetas/fisiologia , Degranulação Celular/fisiologia , Proteína Quinase C/fisiologia , Degranulação Celular/efeitos dos fármacos , Humanos , Isoenzimas/efeitos dos fármacos , Isoenzimas/fisiologia , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Glicoproteínas da Membrana de Plaquetas/fisiologia , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C-delta , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/fisiologia , Transdução de Sinais/efeitos dos fármacos
15.
Curr Pharm Des ; 9(28): 2303-16, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14529392

RESUMO

Platelet P2 receptors--P2Y1, P2Y12, and P2X1--constitute the means by which adenine nucleotides can activate platelets. Coactivation of the Galphaq-coupled P2Y1 and Galphai2-coupled P2Y12 receptors is necessary for ADP-mediated platelet activation, which forms the basis of using P2 antagonists as antithrombotic drugs. P2Y1 receptor antagonists inhibit platelet activation, while P2Y1 knockout mice show longer bleeding times than normal mice but few other problems; however, its ubiquitous expression in other tissues renders P2Y1 questionable as an antithrombotic target. The P2Y12 receptor is expressed nearly exclusively in platelets and brain, making it an attractive antithrombotic target. Antagonists for the P2Y12 receptor have been developed that either require metabolic activation to covalently inhibit P2Y12 and are irreversible, or simply are competitive in nature and thus reversible. Ticlopidine and clopidogrel are irreversible P2Y12 antagonists and have been repeatedly proven as clinical antithrombotic agents. In addition, a recently reported P2Y12 antagonist, CS-747, shows promise as a future antithrombotic drug. The AR-C series of compounds represent reversible P2Y12 antagonists and have been used extensively to characterize the function of P2Y12 in platelets. Clinical studies show that AR-C69931MX is as effective as clopidogrel; furthermore, the combination of AR-C69931MX (cangrelor) and clopidogrel confers greater antagonism of P2Y12 than either antagonist alone. The P2X1 receptor is a calcium channel that functions to potentiate agonist-induced platelet shape change, and its inhibition or loss has little if any effect on hemostasis. A combination of P2Y1 and P2Y12 antagonists may represent an additional course of antithrombotic treatment.


Assuntos
Fibrinolíticos , Antagonistas do Receptor Purinérgico P2 , Animais , Ensaios Clínicos como Assunto , Fibrinolíticos/química , Fibrinolíticos/farmacologia , Fibrinolíticos/uso terapêutico , Humanos , Proteínas de Membrana/antagonistas & inibidores , Estrutura Molecular , Piridinas/química , Piridinas/farmacologia , Piridinas/uso terapêutico , Receptores Purinérgicos P2X , Receptores Purinérgicos P2Y1 , Receptores Purinérgicos P2Y12
16.
Curr Opin Pharmacol ; 3(2): 175-80, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12681240

RESUMO

Activation of P2Y(1) and P2Y(12) receptors, through secreted ADP that is stimulated by agonists such as thrombin, thromboxane and collagen, is a major mechanism of platelet activation. P2X(1) receptors also participate in platelet shape change and potentiation of calcium mobilization. The cloning of the P2Y(12) receptor and its subsequent knockout in mice promises further understanding of its downstream signaling events.


Assuntos
Plaquetas/metabolismo , Ativação Plaquetária/fisiologia , Agregação Plaquetária/fisiologia , Receptores Purinérgicos/fisiologia , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/fisiologia , Humanos , Ativação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Receptores Purinérgicos/química
17.
Blood ; 101(10): 3908-14, 2003 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12560222

RESUMO

Human platelets express 2 G protein-coupled nucleotide receptors: the platelet adenosine diphosphate (ADP) receptor coupled to stimulation of phospholipase C (P2Y(1)) via heterotrimeric guanosine 5-triphosphate (GTP)-binding protein G(q), and the platelet ADP receptor coupled to inhibition of adenylyl cyclase (P2Y(12)) via heterotrimeric GTP-binding protein G(i). Although these 2 receptors are encoded on the same chromosome and have similar pharmacologic profiles, they have different reactivities toward thiol reagents. The thiol agent p-chloromercuribenzene sulfonic acid (pCMBS) and the active metabolites from antiplatelet drugs, clopidogrel and CS-747, inactivate the P2Y(12) receptor and are predicted to interact with the extracellular cysteine residues on the P2Y(12) receptor. In this study we identified the reactive cysteine residues on the human P2Y(12) receptor by site-directed mutagenesis using pCMBS as the thiol reagent. Cys97Ser and Cys175Ser mutants of the P2Y(12) receptor did not express when transfected into Chinese hamster ovary (CHO-K1) cells, indicating the essential nature of a disulfide bridge between these residues. The Cys17Ser, Cys270Ser, and Cys17Ser/Cys270Ser double mutants had similar median effective concentration (EC(50)) values for ADP and 2-methylthio-ADP (2-MeSADP) when compared with the wild-type P2Y(12). Similarly, the median inhibitory concentration (IC(50)) values for BzATP (2',3'-O-(4- benzoylbenzoyl) adenosine 5'-triphosphate), an antagonist of the P2Y(12) receptor, also did not differ dramatically among these mutants and the wild-type P2Y(12) receptor. pCMBS inactivated the wild-type P2Y(12) receptor in a concentration-dependent manner, whereas it had no effect on the P2Y(1) receptor. Finally, pCMBS partially affected the G(i) coupling of Cys17Ser or Cys270Ser receptor mutants, but had no effect on Cys17Ser/Cys270Ser P2Y(12) receptor-mediated inhibition of adenylyl cyclase. These results indicate that, unlike the P2Y(1) receptor, which has 2 essential disulfide bridges linking its extracellular domains, the P2Y(12) receptor has 2 free cysteines in its extracellular domains (Cys17 and Cys270), both of which are targets of thiol reagents. We speculate that the active metabolites of clopidogrel and CS-747 form disulfide bridges with both Cys17 and Cys270 in the P2Y(12) receptor, and thereby inactivate the receptor.


Assuntos
Cisteína , Proteínas de Membrana , Antagonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2/química , Compostos de Sulfidrila/farmacologia , 4-Cloromercuriobenzenossulfonato/farmacologia , Difosfato de Adenosina/farmacologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Células CHO , Cricetinae , AMP Cíclico/metabolismo , Primers do DNA , Humanos , Fosfatos de Inositol/metabolismo , Cinética , Dados de Sequência Molecular , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2Y1 , Receptores Purinérgicos P2Y12 , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transfecção
18.
J Pharmacol Exp Ther ; 303(3): 1114-20, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12438534

RESUMO

Platelet accretion into arterial thrombus in stenotic arterial vessels involves shear-induced platelet activation and adhesion. The Cone and Plate(let) Analyzer (CPA) is designed to simulate such conditions in vitro under a rotating high shear rate in whole blood. In the present study, we evaluated various experimental conditions (including aspirin, temperature, and calcium concentration) and investigated the effects of small molecules along with peptide glycoprotein IIb/IIIa antagonists on platelet adhesion using the CPA system. Concentration-dependent effect of glycoprotein IIb/IIIa antagonists on shear-induced platelet adhesion showed marked differences in potencies: IC50 = 34, 35, 91, 438, and 606 nM for DPC802 (a specific glycoprotein IIb/IIIa antagonist), roxifiban, sibrafiban, lotrafiban, and orbofiban (free acid forms), respectively, and IC50 values of 43, 430, and 5781 nM for abciximab, tirofiban, and eptifibatide, respectively. Parallel study was also conducted to evaluate the effect of glycoprotein IIb/IIIa inhibitors using optical aggregometry. The potency of fibans in blocking shear-induced platelet adhesion correlated well with their binding affinity to the resting and activated glycoprotein IIb/IIIa receptors, as well as their "off-rates". Nevertheless, none of these fibans was able to effectively block shear-induced platelet adhesion at targeted clinical dosing regimens except for abciximab. These data suggest that glycoprotein IIb/IIIa antagonists that show similar efficacy in the inhibition of platelet aggregation in a static in vitro assay may differ substantially in a shear-based system of platelet adhesion. The clinical significance of this phenomenon awaits further investigation.


Assuntos
Ativação Plaquetária/efeitos dos fármacos , Adesividade Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Abciximab , Alanina/metabolismo , Alanina/farmacologia , Amidinas/metabolismo , Amidinas/farmacologia , Análise de Variância , Anticorpos Monoclonais/farmacologia , Anticoagulantes/farmacologia , Aspirina/farmacologia , Agregação Celular/efeitos dos fármacos , Agregação Celular/fisiologia , Relação Dose-Resposta a Droga , Eptifibatida , Humanos , Fragmentos Fab das Imunoglobulinas/farmacologia , Isoxazóis/metabolismo , Isoxazóis/farmacologia , Peptídeos/farmacologia , Ativação Plaquetária/fisiologia , Adesividade Plaquetária/fisiologia , Pirrolidinas/metabolismo , Pirrolidinas/farmacologia , Reologia , Resistência ao Cisalhamento , Temperatura , Tirofibana , Tirosina/análogos & derivados , Tirosina/farmacologia
19.
J Biol Chem ; 277(49): 47588-95, 2002 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-12297512

RESUMO

Activation of GPIIb/IIIa is known to require agonist-induced inside-out signaling through G(q), G(i), and G(z). Although activated by several platelet agonists, including thrombin and thromboxane A(2), the contribution of the G(12/13) signaling pathway to GPIIb/IIIa activation has not been investigated. In this study, we used selective stimulation of G protein pathways to investigate the contribution of G(12/13) activation to platelet fibrinogen receptor activation. YFLLRNP is a PAR-1-specific partial agonist that, at low concentrations (60 microm), selectively activates the G(12/13) signaling cascade resulting in platelet shape change without stimulating the G(q) or G(i) signaling pathways. YFLLRNP-mediated shape change was completely inhibited by the p160(ROCK) inhibitor, Y-27632. At this low concentration, YFLLRNP-mediated G(12/13) signaling caused platelet aggregation and enhanced PAC-1 binding when combined with selective G(i) or G(z) signaling, via selective stimulation of the P2Y(12) receptor or alpha(2A)-adrenergic receptor, respectively. Similar data were obtained when using low dose (10 nm), a thromboxane A(2) mimetic, to activate G(12/13) in the presence of G(i) signaling. These results suggest that selective activation of G(12/13) causes platelet GPIIb/IIIa activation when combined with G(i) signaling. Unlike either G(12/13) or G(i) activation alone, co-activation of both G(12/13) and G(i) resulted in a small increase in intracellular calcium. Chelation of intracellular calcium with dimethyl BAPTA dramatically blocked G(12/13) and G(i)-mediated platelet aggregation. No significant effect on aggregation was seen when using selective inhibitors for p160(ROCK), PKC, or MEKK1. PI 3-kinase inhibition lead to near abolishment of platelet aggregation induced by co-stimulation of G(q) and G(i) pathways, but not by G(12/13) and G(i) pathways. These data demonstrate that co-stimulation of G(12/13) and G(i) pathways is sufficient to activate GPIIb/IIIa in human platelets in a mechanism that involves intracellular calcium, and that PI 3-kinase is an important signaling molecule downstream of G(q) but not downstream of G(12/13) pathway.


Assuntos
Plaquetas/metabolismo , Ácido Egtázico/análogos & derivados , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/química , MAP Quinase Quinase Quinase 1 , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Difosfato de Adenosina/metabolismo , Cálcio/metabolismo , Quelantes/farmacologia , AMP Cíclico/metabolismo , Ácido Egtázico/farmacologia , Eletrofisiologia , Inibidores Enzimáticos/farmacologia , Epinefrina/farmacologia , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Modelos Biológicos , Ligação Proteica , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Tromboxano A2/metabolismo , Quinases Associadas a rho
20.
Biochem J ; 368(Pt 2): 535-43, 2002 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12215172

RESUMO

Platelet fibrinogen receptor activation is a critical step in platelet plug formation. The fibrinogen receptor (integrin alphaIIbbeta3) is activated by agonist-mediated G(q) stimulation and resultant phospholipase C activation. We investigated the role of downstream signalling events from phospholipase C, namely the activation of protein kinase C (PKC) and rise in intracellular calcium, in agonist-induced fibrinogen receptor activation using Ro 31-8220 (a PKC inhibitor) or dimethyl BAPTA [5,5'-dimethyl-bis-(o-aminophenoxy)ethane-N,N,N', N'-tetra-acetic acid], a high-affinity calcium chelator. All the experiments were performed with human platelets treated with aspirin, to avoid positive feedback from thromboxane A2. In the presence of Ro 31-8220, platelet aggregation caused by U46619 was completely inhibited while no effect or partial inhibition was seen with ADP and the thrombin-receptor-activating peptide SFLLRN, respectively. In the presence of intracellular dimethyl BAPTA, ADP- and U46619-induced aggregation and anti-alphaIIbbeta3 antibody PAC-1 binding were completely abolished. However, similar to the effects of Ro 31-8220, dimethyl BAPTA only partially inhibited SFLLRN-induced aggregation, and was accompanied by diminished dense-granule secretion. When either PKC activation or intracellular calcium release was abrogated, aggregation and fibrinogen receptor activation with U46619 or SFLLRN was partially restored by additional selective activation of the G(i) signalling pathway. In contrast, when both PKC activity and intracellular calcium increase were simultaneously inhibited, the complete inhibition of aggregation that occurred in response to either U46619 or SFLLRN could not be restored with concomitant G(i) signalling. We conclude that, while the PKC- and calcium-regulated signalling pathways are capable of inducing activating fibrinogen receptor independently and that each can synergize with G(i) signalling to cause irreversible fibrinogen receptor activation, both pathways act synergistically to effect irreversible fibrinogen receptor activation.


Assuntos
Cálcio/metabolismo , Ácido Egtázico/análogos & derivados , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Fragmentos de Peptídeos/farmacologia , Proteína Quinase C/metabolismo , Receptores de Fibrinogênio/efeitos dos fármacos , Transdução de Sinais , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Difosfato de Adenosina/farmacologia , Plaquetas/citologia , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Células Cultivadas , Fosfatase 2 de Especificidade Dupla , Ácido Egtázico/farmacologia , Inibidores Enzimáticos/farmacologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/efeitos dos fármacos , Humanos , Indóis/farmacologia , Integrina alfa1beta1/metabolismo , Inibidores da Agregação Plaquetária/farmacologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/efeitos dos fármacos , Proteína Fosfatase 2 , Proteínas Tirosina Fosfatases/efeitos dos fármacos , Proteínas Tirosina Fosfatases/metabolismo , Receptores de Fibrinogênio/metabolismo , Tromboxano A2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...