Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Blood ; 123(8): e11-22, 2014 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-24408324

RESUMO

Up to 1% of the population have mild bleeding disorders, but these remain poorly characterized, particularly with regard to the roles of platelets. We have compared the usefulness of Optimul, a 96-well plate-based assay of 7 distinct pathways of platelet activation to characterize inherited platelet defects in comparison with light transmission aggregometry (LTA). Using Optimul and LTA, concentration-response curves were generated for arachidonic acid, ADP, collagen, epinephrine, Thrombin receptor activating-peptide, U46619, and ristocetin in samples from (1) healthy volunteers (n = 50), (2) healthy volunteers treated with antiplatelet agents in vitro (n = 10), and (3) patients with bleeding of unknown origin (n = 65). The assays gave concordant results in 82% of cases (κ = 0.62, P < .0001). Normal platelet function results were particularly predictive (sensitivity, 94%; negative predictive value, 91%), whereas a positive result was not always substantiated by LTA (specificity, 67%; positive predictive value, 77%). The Optimul assay was significantly more sensitive at characterizing defects in the thromboxane pathway, which presented with normal responses with LTA. The Optimul assay is sensitive to mild platelet defects, could be used as a rapid screening assay in patients presenting with bleeding symptoms, and detects changes in platelet function more readily than LTA. This trial was registered at www.isrctn.org as #ISRCTN 77951167.


Assuntos
Transtornos Plaquetários/diagnóstico , Monitoramento de Medicamentos/métodos , Hemorragia/diagnóstico , Ensaios de Triagem em Larga Escala/métodos , Ativação Plaquetária/fisiologia , Inibidores da Agregação Plaquetária/farmacologia , Adulto , Transtornos Plaquetários/sangue , Transtornos Plaquetários/genética , Plaquetas/efeitos dos fármacos , Plaquetas/fisiologia , Feminino , Estudos de Associação Genética , Voluntários Saudáveis , Hemorragia/sangue , Hemorragia/fisiopatologia , Humanos , Masculino , Ativação Plaquetária/efeitos dos fármacos , Valor Preditivo dos Testes , Receptores de Tromboxano A2 e Prostaglandina H2/genética , Sensibilidade e Especificidade , Adulto Jovem
2.
Traffic ; 14(5): 585-98, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23387322

RESUMO

P2Y12 receptor internalization and recycling play an essential role in ADP-induced platelet activation. Recently, we identified a patient with a mild bleeding disorder carrying a heterozygous mutation of P2Y12 (P341A) whose P2Y12 receptor recycling was significantly compromised. Using human cell line models, we identified key proteins regulating wild-type (WT) P2Y12 recycling and investigated P2Y12 -P341A receptor traffic. Treatment with ADP resulted in delayed Rab5-dependent internalization of P341A when compared with WT P2Y12 . While WT P2Y12 rapidly recycled back to the membrane via Rab4 and Rab11 recycling pathways, limited P341A recycling was observed, which relied upon Rab11 activity. Although minimal receptor degradation was evident, P341A was localized in Rab7-positive endosomes with considerable agonist-dependent accumulation in the trans-Golgi network (TGN). Rab7 activity is known to facilitate recruitment of retromer complex proteins to endosomes to transport cargo to the TGN. Here, we identified that P341A colocalized with Vps26; depletion of which blocked limited recycling and promoted receptor degradation. This study has identified key points of divergence in the endocytic traffic of P341A versus WT-P2Y12 . Given that these pathways are retained in human platelets, this research helps define the molecular mechanisms regulating P2Y12 receptor traffic and explain the compromised receptor function in the platelets of the P2Y12 -P341A-expressing patient.


Assuntos
Endossomos/metabolismo , Regulação da Expressão Gênica , Receptores Purinérgicos P2Y12/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Difosfato de Adenosina/metabolismo , Transporte Biológico , Plaquetas/metabolismo , Linhagem Celular , Endocitose , Células HEK293 , Humanos , Ligantes , Proteínas Mutantes/metabolismo , Mutação , Plasmídeos/metabolismo , Estrutura Terciária de Proteína , Proteínas rab de Ligação ao GTP/metabolismo
3.
J Biol Chem ; 287(29): 24505-15, 2012 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-22610101

RESUMO

We have recently shown in a patient with mild bleeding that the PDZ-binding motif of the platelet G protein-coupled P2Y(12) receptor (P2Y(12)R) is required for effective receptor traffic in human platelets. In this study we show for the first time that the PDZ motif-binding protein NHERF1 exerts a major role in potentiating G protein-coupled receptor (GPCR) internalization. NHERF1 interacts with the C-tail of the P2Y(12)R and unlike many other GPCRs, NHERF1 interaction is required for effective P2Y(12)R internalization. In vitro and prior to agonist stimulation P2Y(12)R/NHERF1 interaction requires the intact PDZ binding motif of this receptor. Interestingly on receptor stimulation NHERF1 no longer interacts directly with the receptor but instead binds to the receptor via the endocytic scaffolding protein arrestin. These findings suggest a novel model by which arrestin can serve as an adaptor to promote NHERF1 interaction with a GPCR to facilitate effective NHERF1-dependent receptor internalization.


Assuntos
Arrestina/metabolismo , Fosfoproteínas/metabolismo , Ligação Proteica/fisiologia , Receptores Purinérgicos P2Y12/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Arrestina/genética , Western Blotting , Linhagem Celular , Células Cultivadas , Endocitose , Humanos , Imunoprecipitação , Domínios PDZ/genética , Domínios PDZ/fisiologia , Fosfoproteínas/genética , Ligação Proteica/genética , RNA Interferente Pequeno , Receptores Purinérgicos P2Y12/genética , Trocadores de Sódio-Hidrogênio/genética
4.
Biochem Soc Trans ; 41(1): 225-30, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23356287

RESUMO

Platelets are critical for haemostasis, however inappropriate activation can lead to the development of arterial thrombosis, which can result in heart attack and stroke. ADP is a key platelet agonist that exerts its actions via stimulation of two surface GPCRs (G-protein-coupled receptors), P2Y(1) and P2Y(12). Similar to most GPCRs, P2Y receptor activity is tightly regulated by a number of complex mechanisms including receptor desensitization, internalization and recycling. In the present article, we review the molecular mechanisms that underlie P2Y(1) and P2Y(12) receptor regulation, with particular emphasis on the structural motifs within the P2Y(12) receptor, which are required to maintain regulatory protein interaction. The implications of these findings for platelet responsiveness are also discussed.


Assuntos
Plaquetas/metabolismo , Receptores Purinérgicos P2Y12/fisiologia , Sequência de Aminoácidos , Endocitose , Humanos , Dados de Sequência Molecular , Receptores Purinérgicos P2Y12/química , Receptores Purinérgicos P2Y12/efeitos dos fármacos
5.
Biochem Pharmacol ; 124: 43-56, 2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27845050

RESUMO

Thromboxane A2 is a potent mediator of inflammation and platelet aggregation exerting its effects through the activation of a G protein-coupled receptor (GPCR), termed TP. Although the existence of dimers/oligomers in Class A GPCRs is widely accepted, their functional significance still remains controversial. Recently, we have shown that TPα and TPß homo-/hetero-dimers interact through an interface of residues in transmembrane domain 1 (TM1) whose disruption impairs dimer formation. Here, biochemical and pharmacological characterization of this dimer deficient mutant (DDM) in living cells indicates a significant impairment in its response to agonists. Interestingly, two single loss-of-function TPα variants, namely W29C and N42S recently identified in two heterozygous patients affected by bleeding disorders, match some of the residues mutated in our DDM. These two naturally occurring variants display a reduced potency to TP agonists and are characterized by impaired dimer formation in transfected HEK-293T cells. These findings provide proofs that lack of homo-dimer formation is a crucial process for reduced TPα function in vivo, and might represent one molecular mechanism through which platelet TPα receptor dysfunction affects the patient(s) carrying these mutations.


Assuntos
Plaquetas/fisiologia , Receptores de Tromboxanos/metabolismo , Transdução de Sinais , Dimerização , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Ligantes , Mutação , Receptores de Tromboxanos/agonistas , Receptores de Tromboxanos/antagonistas & inibidores , Receptores de Tromboxanos/genética
6.
PLoS One ; 10(12): e0143913, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26630678

RESUMO

The clinical expression of type 1 von Willebrand disease may be modified by co-inheritance of other mild bleeding diatheses. We previously showed that mutations in the platelet P2Y12 ADP receptor gene (P2RY12) could contribute to the bleeding phenotype in patients with type 1 von Willebrand disease. Here we investigated whether variations in platelet G protein-coupled receptor genes other than P2RY12 also contributed to the bleeding phenotype. Platelet G protein-coupled receptor genes P2RY1, F2R, F2RL3, TBXA2R and PTGIR were sequenced in 146 index cases with type 1 von Willebrand disease and the potential effects of identified single nucleotide variations were assessed using in silico methods and heterologous expression analysis. Seven heterozygous single nucleotide variations were identified in 8 index cases. Two single nucleotide variations were detected in F2R; a novel c.-67G>C transversion which reduced F2R transcriptional activity and a rare c.1063C>T transition predicting a p.L355F substitution which did not interfere with PAR1 expression or signalling. Two synonymous single nucleotide variations were identified in F2RL3 (c.402C>G, p.A134 =; c.1029 G>C p.V343 =), both of which introduced less commonly used codons and were predicted to be deleterious, though neither of them affected PAR4 receptor expression. A third single nucleotide variation in F2RL3 (c.65 C>A; p.T22N) was co-inherited with a synonymous single nucleotide variation in TBXA2R (c.6680 C>T, p.S218 =). Expression and signalling of the p.T22N PAR4 variant was similar to wild-type, while the TBXA2R variation introduced a cryptic splice site that was predicted to cause premature termination of protein translation. The enrichment of single nucleotide variations in G protein-coupled receptor genes among type 1 von Willebrand disease patients supports the view of type 1 von Willebrand disease as a polygenic disorder.


Assuntos
Plaquetas/metabolismo , Receptores Acoplados a Proteínas G/genética , Doenças de von Willebrand/genética , Fator de von Willebrand/genética , Regiões 5' não Traduzidas , Animais , Sequência de Bases , Células HEK293 , Hemorragia/fisiopatologia , Humanos , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único , Homologia de Sequência do Ácido Nucleico , Transcrição Gênica
7.
Thromb Haemost ; 111(5): 923-32, 2014 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-24452735

RESUMO

A small number of thromboxane receptor variants have been described in patients with a bleeding history that result in platelet dysfunction. We have identified a patient with a history of significant bleeding, who expresses a novel heterozygous thromboxane receptor variant that predicts an asparagine to serine substitution (N42S). This asparagine is conserved across all class A GPCRs, suggesting a vital role for receptor structure and function.We investigated the functional consequences of the TP receptor heterozygous N42S substitution by performing platelet function studies on platelet-rich plasma taken from the patient and healthy controls. We investigated the N42S mutation by expressing the wild-type (WT) and mutant receptor in human embryonic kidney (HEK) cells. Aggregation studies showed an ablation of arachidonic acid responses in the patient, whilst there was right-ward shift of the U46619 concentration response curve (CRC). Thromboxane generation was unaffected. Calcium mobilisation studies in cells lines showed a rightward shift of the U46619 CRC in N42S-expressing cells compared to WT. Radioligand binding studies revealed a reduction in BMax in platelets taken from the patient and in N42S-expressing cells, whilst cell studies confirmed poor surface expression. We have identified a novel thromboxane receptor variant, N42S, which results in platelet dysfunction due to reduced surface expression. It is associated with a significant bleeding history in the patient in whom it was identified. This is the first description of a naturally occurring variant that results in the substitution of this highly conserved residue and confirms the importance of this residue for correct GPCR function.


Assuntos
Plaquetas/fisiologia , Hemorragia/genética , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Asparagina/genética , Sequência Conservada/genética , Regulação para Baixo/genética , Feminino , Variação Genética , Células HEK293 , Hemorragia/sangue , Heterozigoto , Humanos , Pessoa de Meia-Idade , Mutação/genética , Ativação Plaquetária/genética , Testes de Função Plaquetária , Ensaio Radioligante , Receptores Acoplados a Proteínas G/genética , Receptores de Tromboxano A2 e Prostaglandina H2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA