Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Int J Mol Sci ; 22(19)2021 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-34638997

RESUMO

One of the mechanisms by which PI3 kinase can regulate platelet function is through phosphorylation of downstream substrates, including glycogen synthase kinase-3 (GSK3)α and GSK3ß. Platelet activation results in the phosphorylation of an N-terminal serine residue in GSK3α (Ser21) and GSK3ß (Ser9), which competitively inhibits substrate phosphorylation. However, the role of phosphorylation of these paralogs is still largely unknown. Here, we employed GSK3α/ß phosphorylation-resistant mouse models to explore the role of this inhibitory phosphorylation in regulating platelet activation. Expression of phosphorylation-resistant GSK3α/ß reduced thrombin-mediated platelet aggregation, integrin αIIbß3 activation, and α-granule secretion, whereas platelet responses to the GPVI agonist collagen-related peptide (CRP-XL) were significantly enhanced. GSK3 single knock-in lines revealed that this divergence is due to differential roles of GSK3α and GSK3ß phosphorylation in regulating platelet function. Expression of phosphorylation-resistant GSK3α resulted in enhanced GPVI-mediated platelet activation, whereas expression of phosphorylation-resistant GSK3ß resulted in a reduction in PAR-mediated platelet activation and impaired in vitro thrombus formation under flow. Interestingly, the latter was normalised in double GSK3α/ß KI mice, indicating that GSK3α KI can compensate for the impairment in thrombosis caused by GSK3ß KI. In conclusion, our data indicate that GSK3α and GSK3ß have differential roles in regulating platelet function.


Assuntos
Plaquetas/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Ativação Plaquetária/genética , Agregação Plaquetária/genética , Transdução de Sinais/genética , Trombose/metabolismo , Animais , Doadores de Sangue , Células Cultivadas , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta/genética , Humanos , Integrinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Trombina/metabolismo , Trombose/genética
2.
J Biol Chem ; 292(5): 1691-1704, 2017 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-27903653

RESUMO

The class I PI3K family of lipid kinases plays an important role in integrin αIIbß3 function, thereby supporting thrombus growth and consolidation. Here, we identify Ras/Rap1GAP Rasa3 (GAP1IP4BP) as a major phosphatidylinositol 3,4,5-trisphosphate-binding protein in human platelets and a key regulator of integrin αIIbß3 outside-in signaling. We demonstrate that cytosolic Rasa3 translocates to the plasma membrane in a PI3K-dependent manner upon activation of human platelets. Expression of wild-type Rasa3 in integrin αIIbß3-expressing CHO cells blocked Rap1 activity and integrin αIIbß3-mediated spreading on fibrinogen. In contrast, Rap1GAP-deficient (P489V) and Ras/Rap1GAP-deficient (R371Q) Rasa3 had no effect. We furthermore show that two Rasa3 mutants (H794L and G125V), which are expressed in different mouse models of thrombocytopenia, lack both Ras and Rap1GAP activity and do not affect integrin αIIbß3-mediated spreading of CHO cells on fibrinogen. Platelets from thrombocytopenic mice expressing GAP-deficient Rasa3 (H794L) show increased spreading on fibrinogen, which in contrast to wild-type platelets is insensitive to PI3K inhibitors. Together, these results support an important role for Rasa3 in PI3K-dependent integrin αIIbß3-mediated outside-in signaling and cell spreading.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais/fisiologia , Substituição de Aminoácidos/genética , Animais , Plaquetas/metabolismo , Plaquetas/patologia , Células CHO , Cricetinae , Cricetulus , Modelos Animais de Doenças , Proteínas Ativadoras de GTPase/genética , Humanos , Camundongos , Camundongos Mutantes , Mutação de Sentido Incorreto , Fosfatidilinositol 3-Quinases/genética , Fosfatos de Fosfatidilinositol/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Receptores Citoplasmáticos e Nucleares/genética , Trombocitopenia/genética , Trombocitopenia/metabolismo , Trombocitopenia/patologia
3.
Blood ; 121(7): 1209-19, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-23243278

RESUMO

Patients with myeloproliferative disorders (MPDs), such as essential thrombocythemia (ET) have increased risk of thrombosis and bleeding, which are major sources of morbidity and mortality. Most MPD patients have a gain of function mutation in Janus kinase 2 (JAK2V617F), but little is known how JAK2V617F affects platelet function. Here, we demonstrate that platelets from ET patients have impaired SFLLRN-mediated fibrinogen binding and have lost the potentiating effect of thrombopoietin (which couples to JAK2) on this pathway. In contrast, SFLLRN-mediated P-selectin expression, ATP secretion, phosphorylation of the PKC substrate pleckstrin, and Ca(2+) mobilization were unaffected in JAK2V617F positive platelets. In addition, thrombopoietin-mediated JAK2 phosphorylation was unchanged, suggesting that signaling pathways activated downstream of JAK2 are impaired. Indeed, we found that platelets from JAK2V617F positive ET patients have significantly reduced phosphorylation of the PI3 kinase substrate Akt, and have reduced activation of Rap1 in response to thrombopoietin, IGF-1,ADP, SFLLRN, and thrombin. This effect was independent of Giα P2Y12 purinergic receptor function as ADP-mediated inhibition of VASP phosphorylation was unchanged. These results demonstrate that the PI3 kinase/Rap1 pathway is intrinsically impaired in platelets from JAK2V617F-positive ET patients, resulting in diminished thrombin and thrombopoietin-mediated integrin α(IIb)ß(3) activation.


Assuntos
Plaquetas/fisiologia , Fosfatidilinositol 3-Quinases/sangue , Ativação Plaquetária/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Proteínas de Ligação a Telômeros/sangue , Trombocitemia Essencial/sangue , Adulto , Idoso , Substituição de Aminoácidos , Plaquetas/efeitos dos fármacos , Estudos de Casos e Controles , Feminino , Fibrinogênio/metabolismo , Humanos , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/sangue , Janus Quinase 2/genética , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Fragmentos de Peptídeos/farmacologia , Fosforilação , Ativação Plaquetária/efeitos dos fármacos , Ativação Plaquetária/genética , Complexo Shelterina , Transdução de Sinais/efeitos dos fármacos , Trombina/farmacologia , Trombocitemia Essencial/genética , Trombopoetina/farmacologia
4.
Arterioscler Thromb Vasc Biol ; 34(8): 1681-8, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24903091

RESUMO

OBJECTIVE: Platelet hyperactivity is a contributing factor in the pathogenesis of cardiovascular disease and can be induced by elevated levels of circulating growth factors, such as insulin-like growth factor-1 (IGF-1). IGF-1 is a primer that cannot stimulate platelet activation by itself, but in combination with physiological stimuli can potentiate platelet functional responses via a phosphoinositide 3-kinase-dependent mechanism. In this study, we explored the role of the phosphoinositide 3-kinase p110α isoform in IGF-1-mediated enhancement of platelet function. APPROACH AND RESULTS: Using a platelet-specific p110α knockout murine model, we demonstrate that genetic deletion, similar to pharmacological inactivation of p110α, did not affect proteinase-activated receptor 4 signaling to Akt/protein kinase B but significantly reduced IGF-1-mediated Akt phosphorylation. The p110ß inhibitor TGX-221 abolished IGF-1-induced Akt phosphorylation in p110α-deficient platelets, demonstrating that both p110α and p110ß contribute to IGF-1-mediated Akt phosphorylation. Genetic deletion of p110α had no effect on IGF-1-mediated increases in thrombus formation on collagen and enhancement of proteinase-activated receptor 4-mediated integrin activation and α-granule secretion. In contrast, pharmacological inhibition of p110α blocked IGF-1-mediated potentiation of integrin activation and α-granule secretion. Functional enhancement by IGF-1 in p110α knockout samples was lost after TGX-221 treatment, suggesting that p110ß drives priming in the absence of the p110α isoform. CONCLUSIONS: Together, these results demonstrate that both p110α and p110ß are involved in Akt signaling by IGF-1, but that it is the p110α isoform that is responsible for IGF-1-mediated potentiation of platelet function.


Assuntos
Plaquetas/enzimologia , Fator de Crescimento Insulin-Like I/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Ativação Plaquetária , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Classe I de Fosfatidilinositol 3-Quinases , Integrina alfa2/metabolismo , Integrina beta3/metabolismo , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/deficiência , Fosfatidilinositol 3-Quinases/genética , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Ativação Plaquetária/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Receptores de Trombina/metabolismo , Vesículas Secretórias/metabolismo , Transdução de Sinais , Trombose/sangue , Trombose/enzimologia , Fatores de Tempo
5.
J Biol Chem ; 288(6): 3918-28, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23239877

RESUMO

Glycogen synthase kinase-3 is a Ser/Thr kinase, tonically active in resting cells but inhibited by phosphorylation of an N-terminal Ser residue (Ser(21) in GSK3α and Ser(9) in GSK3ß) in response to varied external stimuli. Recent work suggests that GSK3 functions as a negative regulator of platelet function, but how GSK3 is regulated in platelets has not been examined in detail. Here, we show that early thrombin-mediated GSK3 phosphorylation (0-30 s) was blocked by PKC inhibitors and largely absent in platelets from PKCα knock-out mice. In contrast, late (2-5 min) GSK3 phosphorylation was dependent on the PI3K/Akt pathway. Similarly, early thrombin-mediated inhibition of GSK3 activity was blocked in PKCα knock-out platelets, whereas the Akt inhibitor MK2206 reduced late thrombin-mediated GSK3 inhibition and largely prevented GSK3 inhibition in PKCα knock-out platelets. More importantly, GSK3 phosphorylation contributes to platelet function as knock-in mice where GSK3α Ser(21) and GSK3ß Ser(9) were mutated to Ala showed a significant reduction in PAR4-mediated platelet aggregation, fibrinogen binding, and P-selectin expression, whereas the GSK3 inhibitor CHIR99021 enhanced these responses. Together, these results demonstrate that PKCα and Akt modulate platelet function by phosphorylating and inhibiting GSK3α/ß, thereby relieving the negative effect of GSK3α/ß on thrombin-mediated platelet activation.


Assuntos
Plaquetas/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Agregação Plaquetária/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Proteína Quinase C-alfa/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Vesículas Secretórias/metabolismo , Trombina/metabolismo , Substituição de Aminoácidos , Animais , Fibrinogênio/metabolismo , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Compostos Heterocíclicos com 3 Anéis/farmacologia , Camundongos , Camundongos Knockout , Mutação de Sentido Incorreto , Selectina-P/biossíntese , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Agregação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Proteína Quinase C-alfa/genética , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Piridinas/farmacologia , Pirimidinas/farmacologia , Vesículas Secretórias/genética , Trombina/farmacologia
6.
BMC Med Genomics ; 16(1): 284, 2023 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-37951941

RESUMO

Deep vein thrombosis (DVT) is the formation of a blood clot in a deep vein. DVT can lead to a venous thromboembolism (VTE), the combined term for DVT and pulmonary embolism, a leading cause of death and disability worldwide. Despite the prevalence and associated morbidity of DVT, the underlying causes are not well understood. Our aim was to leverage publicly available genetic summary association statistics to identify causal risk factors for DVT. We conducted a Mendelian randomization phenome-wide association study (MR-PheWAS) using genetic summary association statistics for 973 exposures and DVT (6,767 cases and 330,392 controls in UK Biobank). There was evidence for a causal effect of 57 exposures on DVT risk, including previously reported risk factors (e.g. body mass index-BMI and height) and novel risk factors (e.g. hyperthyroidism and varicose veins). As the majority of identified risk factors were adiposity-related, we explored the molecular link with DVT by undertaking a two-sample MR mediation analysis of BMI-associated circulating proteins on DVT risk. Our results indicate that circulating neurogenic locus notch homolog protein 1 (NOTCH1), inhibin beta C chain (INHBC) and plasminogen activator inhibitor 1 (PAI-1) influence DVT risk, with PAI-1 mediating the BMI-DVT relationship. Using a phenome-wide approach, we provide putative causal evidence that hyperthyroidism, varicose veins and BMI enhance the risk of DVT. Furthermore, the circulating protein PAI-1 has a causal role in DVT aetiology and is involved in mediating the BMI-DVT relationship.


Assuntos
Hipertireoidismo , Varizes , Trombose Venosa , Humanos , Inibidor 1 de Ativador de Plasminogênio/genética , Fatores de Risco , Trombose Venosa/genética
7.
J Biol Chem ; 286(28): 24553-60, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21592956

RESUMO

Protein kinase B (PKB, Akt) is a Ser/Thr kinase involved in the regulation of cell survival, proliferation, and metabolism and is activated by dual phosphorylation on Thr(308) in the activation loop and Ser(473) in the hydrophobic motif. It plays a contributory role to platelet function, although little is known about its regulation. In this study, we investigated the role of the mammalian target of rapamycin complex (mTORC)-2 in Akt regulation using the recently identified small molecule ATP competitive mTOR inhibitors PP242 and Torin1. Both PP242 and Torin1 blocked thrombin and insulin-like growth factor 1-mediated Akt Ser(473) phosphorylation with an IC(50) between 1 and 5 nm, whereas the mTORC1 inhibitor rapamycin had no effect. Interestingly, PP242 and Torin1 had no effect on Akt Thr(308) phosphorylation, Akt1 activity, and phosphorylation of the Akt substrate glycogen synthase kinase 3ß, indicating that Ser(473) phosphorylation is not necessary for Thr(308) phosphorylation and maximal Akt1 activity. In contrast, Akt2 activity was significantly reduced, concurrent with inhibition of PRAS40 phosphorylation, in the presence of PP242 and Torin1. Other signaling pathways, including phospholipase C/PKC and the MAPK pathway, were unaffected by PP242 and Torin1. Together, these results demonstrate that mTORC2 is the kinase that phosphorylates Akt Ser(473) in human platelets but that this phosphorylation is dispensable for Thr(308) phosphorylation and Akt1 activity.


Assuntos
Plaquetas/enzimologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antibióticos Antineoplásicos/farmacologia , Plaquetas/citologia , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Naftiridinas/farmacologia , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Serina/metabolismo , Sirolimo/farmacologia , Trombina/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fosfolipases Tipo C/metabolismo
8.
J Immunol ; 183(5): 3302-8, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19696433

RESUMO

The phagocytic NADPH oxidase (NOX2) plays a fundamental role in host defense and innate immunity. Here we demonstrate that external ATP triggers rapid cellular oxidation inhibited by diphenyleneiodonium in endotoxin-primed J774 macrophages and primary murine bone marrow-derived macrophages. To identify the source of reactive oxygen species (ROS), we compared responses between wild-type and NOX2-deficient macrophages. ATP-mediated ROS production was strongly attenuated in NOX2-deficient macrophages where responses were comparable to inhibition with diphenyleneiodonium. Notably, spatial differences in superoxide anion formation were observed where ROS formation was partially antagonized by extracellular superoxide dismutase in primary bone marrow-derived macrophages but unaffected in J774 macrophages. Loss of NOX2 was not observed to affect ATP-induced cell death. However, ATP-evoked cell death was found to be partially dependent on caspase-1 and cathepsin B activation. In conclusion, NOX2 plays a fundamental role in conferring macrophages with the ability to respond to extracellular ATP stimulation with robust changes in cellular oxidation.


Assuntos
Trifosfato de Adenosina/fisiologia , Endotoxinas/fisiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Glicoproteínas de Membrana/fisiologia , NADPH Oxidases/fisiologia , Animais , Células da Medula Óssea/enzimologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Linhagem Celular , Células Cultivadas , Líquido Extracelular/enzimologia , Líquido Extracelular/imunologia , Líquido Extracelular/metabolismo , Isoenzimas/deficiência , Isoenzimas/genética , Isoenzimas/fisiologia , Lipopolissacarídeos/fisiologia , Macrófagos/enzimologia , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidase 2 , NADPH Oxidases/deficiência , NADPH Oxidases/genética , Oxirredução , Fagocitose/genética , Fagocitose/imunologia
9.
Sci Rep ; 11(1): 15308, 2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34321503

RESUMO

Sphingosine 1-phosphate (S1P) is a bioactive signalling sphingolipid that is increased in diseases such as obesity and diabetes. S1P can modulate platelet function, however the direction of effect and S1P receptors (S1PRs) involved are controversial. Here we describe the role of S1P in regulating human platelet function and identify the receptor subtypes responsible for S1P priming. Human platelets were treated with protease-activated receptor 1 (PAR-1)-activating peptide in the presence or absence of S1P, S1PR agonists or antagonists, and sphingosine kinases inhibitors. S1P alone did not induce platelet aggregation but at low concentrations S1P enhanced PAR1-mediated platelet responses, whereas PAR1 responses were inhibited by high concentrations of S1P. This biphasic effect was mimicked by pan-S1PR agonists. Specific agonists revealed that S1PR1 receptor activation has a positive priming effect, S1PR2 and S1PR3 have no effect on platelet function, whereas S1PR4 and S1PR5 receptor activation have an inhibitory effect on PAR-1 mediated platelet function. Although platelets express both sphingosine kinase 1/2, enzymes which phosphorylate sphingosine to produce S1P, only dual and SphK2 inhibition reduced platelet function. These results support a role for SphK2-mediated S1P generation in concentration-dependent positive and negative priming of platelet function, through S1PR1 and S1PR4/5 receptors, respectively.


Assuntos
Lisofosfolipídeos/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Receptores de Esfingosina-1-Fosfato/efeitos dos fármacos , Esfingosina/análogos & derivados , Plaquetas/efeitos dos fármacos , Plaquetas/ultraestrutura , Proteínas de Transporte/farmacologia , Forma Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Lisofosfolipídeos/agonistas , Lisofosfolipídeos/antagonistas & inibidores , Fragmentos de Peptídeos/farmacologia , Peptídeos/farmacologia , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Agregação Plaquetária/efeitos dos fármacos , Receptor PAR-1/agonistas , Esfingosina/agonistas , Esfingosina/antagonistas & inibidores , Esfingosina/farmacologia , Receptores de Esfingosina-1-Fosfato/fisiologia
10.
Biochem Pharmacol ; 177: 113975, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32298692

RESUMO

BACKGROUND AND PURPOSE: Rapamycin is a potent immunosuppressant and anti-proliferative agent used clinically to prevent organ transplant rejection and for coating coronary stents to counteract restenosis. Rapamycin complexes with the immunophilin FKBP12, which subsequently binds and inhibits mTORC1. Despite several reports demonstrating that rapamycin affects platelet-mediated responses, the underlying mechanism of how it alters platelet function is poorly characterised. This study aimed to elucidate the effect of rapamycin on platelet procoagulant responses. EXPERIMENTAL APPROACH: The effect of rapamycin on platelet activation and signalling was investigated alongside the catalytic mTOR inhibitors KU0063794 and WYE-687, and the FKBP12-binding macrolide FK506. KEY RESULTS: Rapamycin affects platelet procoagulant responses by reducing externalisation of the procoagulant phospholipid phosphatidylserine, formation of balloon-like structures and local generation of thrombin. Catalytic mTOR kinase inhibitors did not alter platelet procoagulant processes, despite having a similar effect as rapamycin on Ca2+ signalling, demonstrating that the effect of rapamycin on procoagulant responses is independent of mTORC1 inhibition and not linked to a reduction in Ca2+ signalling. FK506, which also forms a complex with FKBP12 but does not target mTOR, reduced platelet procoagulant responses to a similar extent as rapamycin. Both rapamycin and FK506 prevented the loss of mitochondria integrity induced by platelet activation, one of the central regulatory events leading to PS externalisation. CONCLUSIONS AND IMPLICATIONS: Rapamycin suppresses platelet procoagulant responses by protecting mitochondrial integrity in a manner independent of mTORC1 inhibition. Rapamycin and other drugs targeting FKBP immunophilins could aid the development of novel complementary anti-platelet therapies.


Assuntos
Plaquetas/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/genética , Plaquetas/citologia , Plaquetas/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Proteínas de Transporte/farmacologia , Regulação da Expressão Gênica , Humanos , Ionomicina/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Mitocôndrias/metabolismo , Morfolinas/farmacologia , Peptídeos/farmacologia , Fosfatidilserinas/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Cultura Primária de Células , Pirazóis/farmacologia , Pirimidinas/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Tacrolimo/farmacologia , Trombina/metabolismo , Trombina/farmacologia
11.
Cell Signal ; 68: 109528, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31917191

RESUMO

Apoptosis signal-regulating kinase 1 (ASK1) is a member of mitogen-activated protein kinase kinase kinase (MAP3K) family, which recently has been implicated in the regulation of p38 MAPK/PLA2/thromboxane (TxA2) generation, as well as P2Y12 signalling in murine platelets. ASK1 has therefore been proposed as a potential target for anti-thrombotic therapy. At present it is unknown whether ASK1 also contributes to TxA2 formation and platelet function in human. In this study we therefore examined the role of ASK1 using the ASK1 inhibitor selonsertib (GS-4997). We established that ASK1 is responsible for p38 phosphorylation and TxA2 formation in murine platelets, with both GS4997 and p38 inhibitors reducing TxA2 formation. Similar to murine platelets, activation of human platelets resulted in the rapid and transient phosphorylation of ASK1 and the MAP2Ks MMK3/4/6. In contrast, phosphorylation of p38 and its substrate; MAPKAP-kinase2 (MAPKAPK2) was much more sustained. In keeping with these findings, inhibition of ASK1 blocked early, but not later p38/MAPKAPK2 phosphorylation. The latter was dependent on non-canonical autophosphorylation as it was blocked by the p38 inhibitor; SB203580 and the SYK inhibitor; R406. Furthermore, ASK1 and p38 inhibitors had no effect on PLA2 phosphorylation, TxA2 formation and platelet aggregation, demonstrating that this pathway is redundant in human platelets. Together, these results demonstrate that ASK1 contributes to TxA2 formation in murine, but not human platelets and highlight the importance of confirming findings from genetic murine models in humans.


Assuntos
Plaquetas/enzimologia , MAP Quinase Quinase Quinase 5/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Proteína C-Reativa/metabolismo , Colágeno/farmacologia , Ativação Enzimática/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Modelos Biológicos , Fosfolipases A2/metabolismo , Fosforilação/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Glicoproteínas da Membrana de Plaquetas/metabolismo , Tromboxano A2/metabolismo
12.
Artigo em Inglês | MEDLINE | ID: mdl-31743753

RESUMO

After decades in PtdIns(3,4,5)P3's shadow, PtdIns(3,4)P2 has now emerged as a bona fide regulator of important cellular events, including endocytosis and cell migration. New understanding of PtdIns(3,4)P2's cellular roles has been possible via novel approaches to observe and quantify cellular PtdIns(3,4)P2 dynamics, alongside methods to target the kinases and phosphatases governing phosphoinositide turnover. Despite this, the mechanisms by which PtdIns(3,4)P2 orchestrates its cellular roles remain more poorly understood, most notably because, to date, few PtdIns(3,4)P2 effectors have been identified. Here, we develop and apply an affinity-proteomics strategy to conduct a global screen for PtdIns(3,4)P2 interactors in human platelets; a primary cell type with striking PtdIns(3,4)P2 accumulation. Through an integrated approach, coupling affinity capture of PtdIns(3,4)P2-binding proteins to both label-free and isobaric tag-based quantitative proteomics, we identify a diverse PtdIns(3,4)P2 interactome. Included are long-established PtdIns(3,4)P2-binding proteins such as PLEKHA1, PLEKHA2, AKT and DAPP1, and a host of potentially novel effectors, including MTMR5, PNKD, RASA3 and GAB3. The PtdIns(3,4)P2 interactome shows an enrichment of pleckstrin homology (PH) domain-containing proteins, and through bioinformatics and array analyses we characterise the PH domain of MTMR5 and define its phosphoinositide selectivity. The interactome is also diverse in function, including several proteins known to support protein trafficking and cytoskeletal mobilisation. Such proteins have the ability to drive key platelet events, and to fulfil recently-defined roles for PtdIns(3,4)P2 in a wider range of cell types. Moreover, this study will serve as a valuable resource for the future characterisation of effector-driven PtdIns(3,4)P2 function.


Assuntos
Plaquetas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Mapeamento de Interação de Proteínas , Biologia Computacional , Conjuntos de Dados como Assunto , Voluntários Saudáveis , Humanos , Espectrometria de Massas , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapas de Interação de Proteínas , Proteômica
13.
Sci Rep ; 9(1): 1468, 2019 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-30728366

RESUMO

Thrombopoietin (TPO) enhances platelet activation through activation of the tyrosine kinase; JAK2 and the lipid kinase phosphatidylinositide 3-kinase (PI3K). The aim of our study was to identify the PI3K isoforms involved in mediating the effect of TPO on platelet function and elucidate the underlying mechanism. We found that p110ß plays an essential role in TPO-mediated (i) priming of protease-activated receptor (PAR)-mediated integrin αIIbß3 activation and α-granule secretion, (ii) synergistic enhancement of PAR-mediated activation of the small GTPase RAP1, a regulator of integrin activation and (iii) phosphorylation of the PI3K effector Akt. More importantly, the synergistic effect of TPO on phosphorylation of extracellular-regulated kinase (ERK1/2) and thromboxane (TxA2) synthesis was dependent on both p110ß and p110γ. p110ß inhibition/deletion, or inhibition of p110γ, resulted in a partial reduction, whereas inhibiting both p110ß and p110γ completely prevented the synergistic effect of TPO on ERK1/2 phosphorylation and TxA2 synthesis. The latter was ablated by inhibition of MEK, but not p38, confirming a role for ERK1/2 in regulating TPO-mediated increases in TxA2 synthesis. In conclusion, the synergistic effect of TPO on RAP1 and integrin activation is largely mediated by p110ß, whereas p110ß and p110γ contribute to the effect of TPO on ERK1/2 phosphorylation and TxA2 formation.


Assuntos
Plaquetas/metabolismo , Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , Trombopoetina/farmacologia , Tromboxanos/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Humanos , Camundongos , Fosforilação , Ativação Plaquetária/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Tromboxanos/farmacologia , Proteínas rap1 de Ligação ao GTP/metabolismo
14.
Cell Signal ; 19(4): 855-66, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17175137

RESUMO

Non-apoptotic externalization of phosphatidylserine (PS) can act as a reactive surface for the efficient assembly of the prothrombinase complex leading to thrombin generation and coagulation. Here we show that extracellular ATP, acting at the macrophage P2X(7) receptor, drives the rapid Ca(2+)-dependent formation and release of PS-rich microvesicles that enhance the assembly of the prothrombinase complex and subsequent formation of thrombin. Incubation with P2X(7) receptor antagonists (KN-62 and Brilliant Blue G) attenuates ATP induced prothrombotic responses. Consistent with the hypothesis that exposed PS enhances prothrombinase activity; pre-incubation with annexin V blocks the increase in thrombin formation. The rapid translocation of PS and formation of pro-thrombotic microvesicles occurs in the absence of cell lysis. These data demonstrate that the pro-inflammatory P2X(7) receptor can also support and propagate rapid increases in thrombin formation.


Assuntos
Macrófagos/metabolismo , Protrombina/metabolismo , Receptores Purinérgicos P2/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Anexina A5/metabolismo , Transporte Biológico/efeitos dos fármacos , Cálcio/metabolismo , Bovinos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Vesículas Citoplasmáticas/efeitos dos fármacos , Vesículas Citoplasmáticas/metabolismo , Etídio/metabolismo , Humanos , L-Lactato Desidrogenase/metabolismo , Macrófagos/citologia , Macrófagos/ultraestrutura , Camundongos , Fosfatidilserinas/metabolismo , Ligação Proteica/efeitos dos fármacos , Receptores Purinérgicos P2X7 , Tromboplastina/metabolismo
15.
JCI Insight ; 3(10)2018 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-29769447

RESUMO

In response to collagen stimulation, platelets use a coordinated system of fluid entry to undergo membrane ballooning, procoagulant spreading, and microvesiculation. We hypothesized that water entry was mediated by the water channel aquaporin-1 (AQP1) and aimed to determine its role in the platelet procoagulant response and thrombosis. We established that human and mouse platelets express AQP1 and localize to internal tubular membrane structures. However, deletion of AQP1 had minimal effects on collagen-induced platelet granule secretion, aggregation, or membrane ballooning. Conversely, procoagulant spreading, microvesiculation, phosphatidylserine exposure, and clot formation time were significantly diminished. Furthermore, in vivo thrombus formation after FeCl3 injury to carotid arteries was also markedly suppressed in AQP1-null mice, but hemostasis after tail bleeding remained normal. The mechanism involves an AQP1-mediated rapid membrane stretching during procoagulant spreading but not ballooning, leading to calcium entry through mechanosensitive cation channels and a full procoagulant response. We conclude that AQP1 is a major regulator of the platelet procoagulant response, able to modulate coagulation after injury or pathologic stimuli without affecting other platelet functional responses or normal hemostasis. Clinically effective AQP1 inhibitors may therefore represent a novel class of antiprocoagulant antithrombotics.


Assuntos
Aquaporina 1/fisiologia , Plaquetas/metabolismo , Coagulantes/metabolismo , Trombose/fisiopatologia , Animais , Aquaporina 1/antagonistas & inibidores , Aquaporina 1/genética , Aquaporina 1/metabolismo , Membrana Celular/metabolismo , Humanos , Camundongos , Camundongos Knockout , Frações Subcelulares/metabolismo
17.
Blood Adv ; 1(14): 918-932, 2017 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-29242851

RESUMO

The class I phosphoinositide 3-kinase (PI3K) isoforms play important roles in platelet priming, activation, and stable thrombus formation. Class I PI3Ks predominantly regulate cell function through their catalytic product, the signaling phospholipid phosphatidylinositol 3,4,5-trisphosphate [PtdIns(3,4,5)P3], which coordinates the localization and/or activity of a diverse range of binding proteins. Notably, the complete repertoire of these class I PI3K effectors in platelets remains unknown, limiting mechanistic understanding of class I PI3K-mediated control of platelet function. We measured robust agonist-driven PtdIns (3,4,5)P3 generation in human platelets by lipidomic mass spectrometry (MS), and then used affinity-capture coupled to high-resolution proteomic MS to identify the targets of PtdIns (3,4,5)P3 in these cells. We reveal for the first time a diverse platelet PtdIns(3,4,5)P3 interactome, including kinases, signaling adaptors, and regulators of small GTPases, many of which are previously uncharacterized in this cell type. Of these, we show dual adaptor for phosphotyrosine and 3-phosphoinositides (DAPP1) to be regulated by Src-family kinases and PI3K, while platelets from DAPP1-deficient mice display enhanced thrombus formation on collagen in vitro. This was associated with enhanced platelet α/δ granule secretion and αIIbß3 integrin activation downstream of the collagen receptor glycoprotein VI. Thus, we present the first comprehensive analysis of the PtdIns(3,4,5)P3 signalosome of human platelets and identify DAPP1 as a novel negative regulator of platelet function. This work provides important new insights into how class I PI3Ks shape platelet function.

18.
Cardiovasc Res ; 107(1): 9-19, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25902782

RESUMO

AIMS: Patients with conditions that are associated with insulin resistance such as obesity, type 2 diabetes mellitus, and polycystic ovary syndrome have an increased risk of thrombosis and a concurrent hyperactive platelet phenotype. Our aim was to determine whether insulin resistance of megakaryocytes/platelets promotes platelet hyperactivation. METHODS AND RESULTS: We generated a conditional mouse model where the insulin receptor (IR) was specifically knocked out in megakaryocytes/platelets and performed ex vivo platelet activation studies in wild-type (WT) and IR-deficient platelets by measuring aggregation, integrin αIIbß3 activation, and dense and α-granule secretion. Deletion of IR resulted in an increase in platelet count and volume, and blocked the action of insulin on platelet signalling and function. Platelet aggregation, granule secretion, and integrin αIIbß3 activation in response to the glycoprotein VI (GPVI) agonist collagen-related peptide (CRP) were significantly reduced in platelets lacking IR. This was accompanied by a reduction in the phosphorylation of effectors downstream of GPVI. Interestingly, loss of IR also resulted in a reduction in insulin-like growth factor-1 (IGF-1)- and insulin-like growth factor-2 (IGF-2)-mediated phosphorylation of IRS-1, Akt, and GSK3ß and priming of CRP-mediated platelet activation. Pharmacological inhibition of IR and the IGF-1 receptor in WT platelets recapitulated the platelet phenotype of IR-deficient platelets. CONCLUSIONS: Deletion of IR (i) increases platelet count and volume, (ii) does not cause platelet hyperactivity, and (iii) reduces GPVI-mediated platelet function and platelet priming by IGF-1 and IGF-2.


Assuntos
Plaquetas/fisiologia , Fator de Crescimento Insulin-Like II/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Megacariócitos/fisiologia , Receptor de Insulina/fisiologia , Transdução de Sinais/fisiologia , Trombocitose/etiologia , Animais , Insulina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Glicoproteínas da Membrana de Plaquetas/fisiologia
20.
Biochem Pharmacol ; 76(12): 1740-7, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18848528

RESUMO

Zinc (Zn2+) and copper (Cu2+) are key signalling molecules in the immune system and regulate the activity of many ion channels. Both Zn2+ and Cu2+ potently inhibit rat P2X7 receptors via a binding site identified by mutagenesis. Here we show that extracellular Cu2+ also potently inhibits mouse P2X7 receptors. By contrast, the receptor expression system and agonist strongly influence the action of extracellular Zn2+ at mouse P2X7 receptors. Consistent with previous reports, Zn2+ inhibits recombinant rat P2X7 receptors. However, recombinant mouse P2X7 receptors are potentiated by Zn2+ when activated by ATP4- but inhibited when stimulated with the ATP analogue BzATP4-. Endogenous murine macrophage P2X7 receptors are not modulated by Zn2+ when stimulated by ATP4- however Zn2+ inhibits BzATP4- mediated responses. In summary, these findings provide a fundamental insight into the differential actions of Zn2+ and Cu2+ between different P2X7 receptor species.


Assuntos
Receptores Purinérgicos P2/efeitos dos fármacos , Zinco/farmacologia , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Animais , Cobre/farmacologia , Macrófagos/química , Camundongos , Agonistas do Receptor Purinérgico P2 , Ratos , Receptores Purinérgicos P2X7 , Proteínas Recombinantes , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA