Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
BMC Biol ; 21(1): 14, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36721118

RESUMEN

BACKGROUND: Purinergic P2Y1 and P2Y12 receptors (P2Y1-R and P2Y12-R) are G protein-coupled receptors (GPCR) activated by adenosine diphosphate (ADP) to mediate platelet activation, thereby playing a pivotal role in hemostasis and thrombosis. While P2Y12-R is the major target of antiplatelet drugs, no P2Y1-R antagonist has yet been developed for clinical use. However, accumulating data suggest that P2Y1-R inhibition would ensure efficient platelet inhibition with minimal effects on bleeding. In this context, an accurate characterization of P2Y1-R antagonists constitutes an important preliminary step. RESULTS: Here, we investigated the pharmacology of P2Y1-R signaling through Gq and ß-arrestin pathways in HEK293T cells and in mouse and human platelets using highly sensitive resonance energy transfer-based technologies (BRET/HTRF). We demonstrated that at basal state, in the absence of agonist ligand, P2Y1-R activates Gq protein signaling in HEK293T cells and in mouse and human platelets, indicating that P2Y1-R is constitutively active in physiological conditions. We showed that P2Y1-R also promotes constitutive recruitment of ß-arrestin 2 in HEK293T cells. Moreover, the P2Y1-R antagonists MRS2179, MRS2279 and MRS2500 abolished the receptor dependent-constitutive activation, thus behaving as inverse agonists. CONCLUSIONS: This study sheds new light on P2Y1-R pharmacology, highlighting for the first time the existence of a constitutively active P2Y1-R population in human platelets. Given the recent interest of P2Y12-R constitutive activity in patients with diabetes, this study suggests that modification of constitutive P2Y1-R signaling might be involved in pathological conditions, including bleeding syndrome or high susceptibility to thrombotic risk. Thus, targeting platelet P2Y1-R constitutive activation might be a promising and powerful strategy for future antiplatelet therapy.


Asunto(s)
Agonismo Inverso de Drogas , Proteínas de Unión al GTP , Receptores Purinérgicos P2Y1 , Transducción de Señal , Arrestina beta 2 , Animales , Humanos , Ratones , Arrestina beta 2/metabolismo , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Receptores Purinérgicos P2Y1/metabolismo , Plaquetas
2.
EMBO Rep ; 22(6): e51299, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-33880878

RESUMEN

Endothelium protection is critical, because of the impact of vascular leakage and edema on pathological conditions such as brain ischemia. Whereas deficiency of class II phosphoinositide 3-kinase alpha (PI3KC2α) results in an increase in vascular permeability, we uncover a crucial role of the beta isoform (PI3KC2ß) in the loss of endothelial barrier integrity following injury. Here, we studied the role of PI3KC2ß in endothelial permeability and endosomal trafficking in vitro and in vivo in ischemic stroke. Mice with inactive PI3KC2ß showed protection against vascular permeability, edema, cerebral infarction, and deleterious inflammatory response. Loss of PI3KC2ß in human cerebral microvascular endothelial cells stabilized homotypic cell-cell junctions by increasing Rab11-dependent VE-cadherin recycling. These results identify PI3KC2ß as a potential new therapeutic target to prevent aggravating lesions following ischemic stroke.


Asunto(s)
Células Endoteliales , Fosfatidilinositol 3-Quinasas , Uniones Adherentes/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Permeabilidad Capilar , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Ratones , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo
3.
Int J Mol Sci ; 24(2)2023 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-36674478

RESUMEN

The Src homology 2 domain-containing inositol 5-phosphatase 1 (SHIP1) is known to dephosphorylate PtdIns(3,4,5)P3 into PtdIns(3,4)P2 and to interact with several signaling proteins though its docking functions. It has been shown to negatively regulate platelet adhesion and spreading on a fibrinogen surface and to positively regulate thrombus growth. In the present study, we have investigated its role during the early phase of platelet activation. Using confocal-based morphometric analysis, we found that SHIP1 is involved in the regulation of cytoskeletal organization and internal contractile activity in thrombin-activated platelets. The absence of SHIP1 has no significant impact on thrombin-induced Akt or Erk1/2 activation, but it selectively affects the RhoA/Rho-kinase pathway and myosin IIA relocalization to the cytoskeleton. SHIP1 interacts with the spectrin-based membrane skeleton, and its absence induces a loss of sustained association of integrins to this network together with a decrease in αIIbß3 integrin clustering following thrombin stimulation. This αIIbß3 integrin dynamics requires the contractile cytoskeleton under the control of SHIP1. RhoA activation, internal platelet contraction, and membrane skeleton integrin association were insensitive to the inhibition of PtdIns(3,4,5)P3 synthesis or SHIP1 phosphatase activity, indicating a role for the docking properties of SHIP1 in these processes. Altogether, our data reveal a lipid-independent function for SHIP1 in the regulation of the contractile cytoskeleton and integrin dynamics in platelets.


Asunto(s)
Integrina alfa2 , Integrina beta3 , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Activación Plaquetaria , Plaquetas/metabolismo , Integrina beta3/metabolismo , Fosfatidilinositoles/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Trombina/farmacología , Trombina/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/metabolismo , Integrina alfa2/metabolismo
4.
Platelets ; 33(6): 918-925, 2022 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-34915822

RESUMEN

During severe sepsis, platelet activation may induce disseminate microvascular thrombosis, which play a key role in critical organ failure. Crucially, most of the studies in this field have explored platelet-leukocyte interactions in animal models, or explored platelets under the spectrum of thrombocytopenia or disseminated intravascular coagulation and have not taken into account the complex interplay that might exist between platelets and leukocytes during human septic shock nor the kinetics of platelet activation. Here, we assessed platelet activation parameters at the admission of patients with sepsis to the intensive care unit (ICU) and 48 hours later. Twenty-two patients were enrolled in the study, thirteen (59.1%) of whom were thrombocytopenic. The control group was composed of twelve infection-free patients admitted during the study period. The activation parameters studied included platelet-leukocyte interactions, assessed by flow cytometry in whole blood, as well as membrane surface and soluble platelet activation markers measured by flow cytometry and dedicated ELISA kits. We also investigated platelet aggregation and secretion responses of patients with sepsis following stimulation, compared to controls. At admission, the level of circulating monocyte-platelet and neutrophil-platelet heterotypic aggregates was significantly higher in sepsis patients compared to controls and returned to a level comparable to controls or even below 48 hours later. Basal levels of CD62P and CD63 platelet membrane exposure at admission and 48 hours later were low and similar to controls. In contrast, plasma level of soluble GPVI and soluble CD40 ligand was significantly increased in septic patients, at the two times of analysis, reflecting previous platelet activation. Platelet aggregation and secretion responses induced by specific agonists were significantly decreased in septic conditions, particularly 48 hours after admission. Hence, we have observed for the first time that critically ill septic patients compared to controls have both an early and durable platelet activation while their circulating platelets are less responsive to different agonists.


Asunto(s)
Sepsis , Choque Séptico , Animales , Plaquetas/fisiología , Humanos , Unidades de Cuidados Intensivos , Activación Plaquetaria/fisiología
5.
Biochem J ; 478(6): 1199-1225, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33740047

RESUMEN

PI3Ks are important lipid kinases that produce phosphoinositides phosphorylated in position 3 of the inositol ring. There are three classes of PI3Ks: class I PI3Ks produce PIP3 at plasma membrane level. Although D. melanogaster and C. elegans have only one form of class I PI3K, vertebrates have four class I PI3Ks called isoforms despite being encoded by four different genes. Hence, duplication of these genes coincides with the acquisition of coordinated multi-organ development. Of the class I PI3Ks, PI3Kα and PI3Kß, encoded by PIK3CA and PIK3CB, are ubiquitously expressed. They present similar putative protein domains and share PI(4,5)P2 lipid substrate specificity. Fifteen years after publication of their first isoform-selective pharmacological inhibitors and genetically engineered mouse models (GEMMs) that mimic their complete and specific pharmacological inhibition, we review the knowledge gathered in relation to the redundant and selective roles of PI3Kα and PI3Kß. Recent data suggest that, further to their redundancy, they cooperate for the integration of organ-specific and context-specific signal cues, to orchestrate organ development, physiology, and disease. This knowledge reinforces the importance of isoform-selective inhibitors in clinical settings.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Fosfatidilinositoles/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Animales , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Humanos , Fosforilación , Transducción de Señal , Especificidad por Sustrato
6.
Br J Haematol ; 192(5): 909-921, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33528045

RESUMEN

Lowe syndrome (LS) is an oculocerebrorenal syndrome of Lowe (OCRL1) genetic disorder resulting in a defect of the OCRL protein, a phosphatidylinositol-4,5-bisphosphate 5-phosphatase containing various domains including a Rho GTPase-activating protein (RhoGAP) homology domain catalytically inactive. We previously reported surgery-associated bleeding in patients with LS, suggestive of platelet dysfunction, accompanied with a mild thrombocytopenia in several patients. To decipher the role of OCRL in platelet functions and in megakaryocyte (MK) maturation, we conducted a case-control study on 15 patients with LS (NCT01314560). While all had a drastically reduced expression of OCRL, this deficiency did not affect platelet aggregability, but resulted in delayed thrombus formation on collagen under flow conditions, defective platelet spreading on fibrinogen and impaired clot retraction. We evidenced alterations of the myosin light chain phosphorylation (P-MLC), with defective Rac1 activity and, inversely, elevated active RhoA. Altered cytoskeleton dynamics was also observed in cultured patient MKs showing deficient proplatelet extension with increased P-MLC that was confirmed using control MKs transfected with OCRL-specific small interfering(si)RNA (siOCRL). Patients with LS also had an increased proportion of circulating barbell-shaped proplatelets. Our present study establishes that a deficiency of the OCRL protein results in a defective actomyosin cytoskeleton reorganisation in both MKs and platelets, altering both thrombopoiesis and some platelet responses to activation necessary to ensure haemostasis.


Asunto(s)
Plaquetas/citología , Megacariocitos/citología , Síndrome Oculocerebrorrenal/genética , Monoéster Fosfórico Hidrolasas/fisiología , Trombopoyesis/fisiología , Actomiosina/análisis , Adolescente , Adulto , Anemia/etiología , Coagulación Sanguínea , Plaquetas/ultraestructura , Estudios de Casos y Controles , Forma de la Célula , Niño , Colágeno , Citoesqueleto/ultraestructura , Femenino , Silenciador del Gen , Humanos , Masculino , Megacariocitos/ultraestructura , Persona de Mediana Edad , Mutación , Cadenas Ligeras de Miosina/metabolismo , Síndrome Oculocerebrorrenal/sangre , Síndrome Oculocerebrorrenal/patología , Monoéster Fosfórico Hidrolasas/deficiencia , Monoéster Fosfórico Hidrolasas/genética , Fosforilación , Dominios Proteicos , Procesamiento Proteico-Postraduccional , ARN Interferente Pequeño/genética , Transducción de Señal , Trombocitopenia/etiología , Adulto Joven
7.
Biochem J ; 477(22): 4327-4342, 2020 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-33242335

RESUMEN

Our knowledge on the expression, regulation and roles of the different phosphoinositide 3-kinases (PI3Ks) in platelet signaling and functions has greatly expanded these last twenty years. Much progress has been made in understanding the roles and regulations of class I PI3Ks which produce the lipid second messenger phosphatidylinositol 3,4,5 trisphosphate (PtdIns(3,4,5)P3). Selective pharmacological inhibitors and genetic approaches have allowed researchers to generate an impressive amount of data on the role of class I PI3Kα, ß, δ and γ in platelet activation and in thrombosis. Furthermore, platelets do also express two class II PI3Ks (PI3KC2α and PI3KC2ß), thought to generate PtdIns(3,4)P2 and PtdIns3P, and the sole class III PI3K (Vps34), known to synthesize PtdIns3P. Recent studies have started to reveal the importance of PI3KC2α and Vps34 in megakaryocytes and platelets, opening new perspective in our comprehension of platelet biology and thrombosis. In this review, we will summarize previous and recent advances on platelet PI3Ks isoforms. The implication of these kinases and their lipid products in fundamental platelet biological processes and thrombosis will be discussed. Finally, the relevance of developing potential antithrombotic strategies by targeting PI3Ks will be examined.


Asunto(s)
Plaquetas/enzimología , Fosfatidilinositol 3-Quinasas Clase II/metabolismo , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Trombosis/enzimología , Trombosis/terapia , Animales , Plaquetas/patología , Humanos , Isoenzimas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Trombosis/patología
8.
Cancer Metastasis Rev ; 37(2-3): 477-489, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29868934

RESUMEN

Our knowledge on the role of the different lipid messengers produced by phosphoinositide 3-kinases (PI3Ks) in normal and cancer cells as well as in platelets during arterial thrombosis has greatly expanded these last 15 years. PI3Ks are a family of lipid kinases that catalyze the phosphorylation of the D3 position of the inositol ring of phosphoinositides to produce phosphatidylinositol 3-phosphate (PtdIns3P), phosphatidylinositol 3,4-bisphosphate (PtdIns(3,4)P2), and phosphatidylinositol-3,4,5 trisphosphate (PtdIns(3,4,5)P3). These D3-phosphoinositides act as intracellular messengers recruiting effector proteins involved in the control of diverse cellular functions including survival, proliferation, migration, membrane trafficking, and cytoskeleton dynamics. The current idea is that the different isoforms of PI3Ks produce specific pools of lipids that regulate in time and space, at the membrane/cytosol interface, the formation of appropriate functional protein complexes. Dysregulation of PI3K-dependent pathways is directly involved in the etiology of several pathologies including cancers where the PI3K/AKT/mTORC1 axis is frequently aberrantly activated. Moreover, PtdIns(3,4,5)P3 production has been shown to play an essential role in platelet functions, particularly in the formation of a stable platelet thrombus at high shear rate. Therefore, PI3Ks are attractive therapeutic targets in the treatment of cancer and arterial thrombosis. In this review, we will discuss the role of the different lipid products of PI3K isoforms in the context of cancer and thrombosis and the development of selective PI3Ks inhibitors in the treatment of these diseases.


Asunto(s)
Metabolismo de los Lípidos , Neoplasias/etiología , Neoplasias/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Trombosis/etiología , Trombosis/metabolismo , Animales , Plaquetas/metabolismo , Membrana Celular/metabolismo , Homeostasis , Humanos , Isoenzimas , Fosfatidilinositol 3-Quinasas/química , Fosfatidilinositoles/química , Fosfatidilinositoles/metabolismo , Transducción de Señal , Relación Estructura-Actividad
9.
Blood ; 130(18): 2032-2042, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-28903944

RESUMEN

To uncover the role of Vps34, the sole class III phosphoinositide 3-kinase (PI3K), in megakaryocytes (MKs) and platelets, we created a mouse model with Vps34 deletion in the MK/platelet lineage (Pf4-Cre/Vps34lox/lox). Deletion of Vps34 in MKs led to the loss of its regulator protein, Vps15, and was associated with microthrombocytopenia and platelet granule abnormalities. Although Vps34 deficiency did not affect MK polyploidisation or proplatelet formation, it dampened MK granule biogenesis and directional migration toward an SDF1α gradient, leading to ectopic platelet release within the bone marrow. In MKs, the level of phosphatidylinositol 3-monophosphate (PI3P) was significantly reduced by Vps34 deletion, resulting in endocytic/trafficking defects. In platelets, the basal level of PI3P was only slightly affected by Vps34 loss, whereas the stimulation-dependent pool of PI3P was significantly decreased. Accordingly, a significant increase in the specific activity of Vps34 lipid kinase was observed after acute platelet stimulation. Similar to Vps34-deficient platelets, ex vivo treatment of wild-type mouse or human platelets with the Vps34-specific inhibitors, SAR405 and VPS34-IN1, induced abnormal secretion and affected thrombus growth at arterial shear rate, indicating a role for Vps34 kinase activity in platelet activation, independent from its role in MKs. In vivo, Vps34 deficiency had no impact on tail bleeding time, but significantly reduced platelet prothrombotic capacity after carotid injury. This study uncovers a dual role for Vps34 as a regulator of platelet production by MKs and as an unexpected regulator of platelet activation and arterial thrombus formation dynamics.


Asunto(s)
Plaquetas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Trombosis/enzimología , Trombosis/patología , Animales , Linaje de la Célula , Movimiento Celular , Gránulos Citoplasmáticos/metabolismo , Espacio Intracelular/metabolismo , Megacariocitos/metabolismo , Megacariocitos/ultraestructura , Ratones Endogámicos C57BL , Fosfatos de Fosfatidilinositol/metabolismo , Transporte de Proteínas , Reproducibilidad de los Resultados , Trombocitopenia/patología
10.
Arterioscler Thromb Vasc Biol ; 38(9): 2041-2053, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30354258

RESUMEN

Objective- PI3Kα (phosphoinositide 3-kinase alpha) is a therapeutic target in oncology, but its role in platelets and thrombosis remains ill characterized. In this study, we have analyzed the role of PI3Kα in vitro, ex vivo, and in vivo in 2 models of arterial thrombosis. Approach and Results- Using mice selectively deficient in p110α in the megakaryocyte lineage and isoform-selective inhibitors, we confirm that PI3Kα is not mandatory but participates to thrombus growth over a collagen matrix at arterial shear rate. Our data uncover a role for PI3Kα in low-level activation of the GP (glycoprotein) VI-collagen receptor by contributing to ADP secretion and in turn full activation of PI3Kß and Akt/PKB (protein kinase B). This effect was no longer observed at high level of GP VI agonist concentration. Our study also reveals that over a vWF (von Willebrand factor) matrix, PI3Kα regulates platelet stationary adhesion contacts under arterial flow through its involvement in the outside-in signaling of vWF-engaged αIIbß3 integrin. In vivo, absence or inhibition of PI3Kα resulted in a modest but significant decrease in thrombus size after superficial injuries of mouse mesenteric arteries and an increased time to arterial occlusion after carotid lesion, without modification in the tail bleeding time. Considering the more discrete and nonredundant role of PI3Kα compared with PI3Kß, selective PI3Kα inhibitors are unlikely to increase the bleeding risk at least in the absence of combination with antiplatelet drugs or thrombopenia. Conclusions- This study provides mechanistic insight into the role of PI3Kα in platelet activation and arterial thrombosis.


Asunto(s)
Hemostasis , Fosfatidilinositol 3-Quinasa/fisiología , Adhesividad Plaquetaria , Agregación Plaquetaria , Trombosis/fisiopatología , Animales , Activación Enzimática , Femenino , Humanos , Masculino , Ratones , Inhibidores de las Quinasa Fosfoinosítidos-3 , Glicoproteínas de Membrana Plaquetaria/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factor de von Willebrand/metabolismo
11.
Int J Mol Sci ; 20(14)2019 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-31315248

RESUMEN

Host defense against infection is based on two crucial mechanisms: the inflammatory response and the activation of coagulation. Platelets are involved in both hemostasis and immune response. These mechanisms work together in a complex and synchronous manner making the contribution of platelets of major importance in sepsis. This is a summary of the pathophysiology of sepsis-induced thrombocytopenia, microvascular consequences, platelet-endothelial cells and platelet-pathogens interactions. The critical role of platelets during sepsis and the therapeutic implications are also reviewed.


Asunto(s)
Plaquetas/inmunología , Sepsis/sangre , Trombocitopenia/sangre , Animales , Toxinas Bacterianas/toxicidad , Plaquetas/patología , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Humanos , Sepsis/complicaciones , Trombocitopenia/etiología
12.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(9): 1121-1131, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29902570

RESUMEN

Phosphoinositides are bioactive lipids essential in the regulation of cell signaling as well as cytoskeleton and membrane dynamics. Their metabolism is highly active in blood platelets where they play a critical role during activation, at least through two well identified pathways involving phospholipase C and phosphoinositide 3-kinases (PI3K). Here, using a sensitive high-performance liquid chromatography-mass spectrometry method recently developed, we monitored for the first time the profiling of phosphatidylinositol (PI), PIP, PIP2 and PIP3 molecular species (fatty-acyl profiles) in human and mouse platelets during the course of stimulation by thrombin and collagen-related peptide. Furthermore, using class IA PI3K p110α or p110ß deficient mouse platelets and a pharmacological inhibitor, we show the crucial role of p110ß and the more subtle role of p110α in the production of PIP3 molecular species following stimulation. This comprehensive platelet phosphoinositides profiling provides important resources for future studies and reveals new information on phosphoinositides biology, similarities and differences in mouse and human platelets and unexpected dramatic increase in low-abundance molecular species of PIP2 during stimulation, opening new perspectives in phosphoinositide signaling in platelets.


Asunto(s)
Plaquetas/efectos de los fármacos , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Animales , Plaquetas/citología , Plaquetas/metabolismo , Proteínas Portadoras/farmacología , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasa Clase I/deficiencia , Inhibidores Enzimáticos/farmacología , Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos/farmacología , Activación Plaquetaria/efectos de los fármacos , Cultivo Primario de Células , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Pirimidinonas/farmacología , Trombina/farmacología , ortoaminobenzoatos/farmacología
13.
Blood ; 125(5): 881-8, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25398937

RESUMEN

Class IA phosphoinositide 3-kinase ß (PI3Kß) is considered a potential drug target in arterial thrombosis, which is a major cause of death worldwide. Here we show that a striking phenotype of mice with selective p110ß deletion in the megakaryocyte lineage is thrombus instability at a high shear rate, which is an effect that is not detected in the absence of p110α in platelets. The high shear rate-dependent thrombus instability in the absence of p110ß is observed both ex vivo and in vivo with the formation of platelet emboli. Moreover, PI3Kß is required for the recruitment of new platelets to a growing thrombus when a pathological high shear is applied. Treatment of human blood with AZD6482, a selective PI3Kß inhibitor, phenocopies p110ß deletion in mouse platelets, which highlights the role of the kinase activity of p110ß. Within the growing platelet thrombus, p110ß inactivation impairs the activating phosphorylations of Akt and the inhibitory phosphorylation of GSK3. In accord with these data, pharmacologic inhibition of GSK3 restores thrombus stability. Thus, platelet PI3Kß is not essential for thrombus growth and stability at normal arterial shear but has a specific and critical role in maintaining the integrity of the formed thrombus on elevation of shear rate, suggesting a potential risk of embolization on treatment with PI3Kß inhibitors.


Asunto(s)
Plaquetas/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/genética , Glucógeno Sintasa Quinasa 3/genética , Trombosis/genética , Animales , Plaquetas/efectos de los fármacos , Plaquetas/patología , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasa Clase I/deficiencia , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Mecanotransducción Celular , Ratones , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Pirimidinonas/farmacología , Estrés Mecánico , Trombosis/enzimología , Trombosis/patología , ortoaminobenzoatos/farmacología
14.
Blood ; 126(9): 1128-37, 2015 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-26109204

RESUMEN

The physiologic roles of the class II phosphoinositide 3-kinases (PI3Ks) and their contributions to phosphatidylinositol 3-monophosphate (PI3P) and PI(3,4)P2 production remain elusive. Here we report that mice heterozygous for a constitutively kinase-dead PI3K-C2α display aberrant platelet morphology with an elevated number of barbell-shaped proplatelets, a recently discovered intermediate stage in the final process of platelet production. Platelets with heterozygous PI3K-C2α inactivation have critical defects in α-granules and membrane structure that are associated with modifications in megakaryocytes. These platelets are more rigid and unable to form filopodia after stimulation. Heterozygous PI3K-C2α inactivation in platelets led to a significant reduction in the basal pool of PI3P and a mislocalization of several membrane skeleton proteins known to control the interactions between the plasma membrane and cytoskeleton. These alterations had repercussions on the performance of platelet responses with delay in the time of arterial occlusion in an in vivo model of thrombosis and defect in thrombus formation in an ex vivo blood flow system. These data uncover a key role for PI3K-C2α activity in the generation of a basal housekeeping PI3P pool and in the control of membrane remodeling, critical for megakaryocytopoiesis and normal platelet production and function.


Asunto(s)
Plaquetas/patología , Membrana Celular/patología , Mutación , Fosfatidilinositol 3-Quinasas/genética , Animales , Plaquetas/citología , Plaquetas/metabolismo , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Técnicas de Sustitución del Gen , Heterocigoto , Metabolismo de los Lípidos , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Trombopoyesis
15.
Biochem Biophys Res Commun ; 476(3): 167-73, 2016 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-27155155

RESUMEN

Phosphoinositides play a key role in the spatiotemporal control of central intracellular processes and several specific kinases and phosphatases regulating the level of these lipids are implicated in human diseases. Myotubularins are a family of 3-phosphatases acting specifically on phosphatidylinositol 3-monophosphate and phosphatidylinositol 3,5 bisphosphate. Members of this family are mutated in genetic diseases including myotubularin 1 (MTM1) and myotubularin-related protein 2 (MTMR2) which mutations are responsible of X-linked centronuclear myopathy and Charcot-Marie-Tooth neuropathy, respectively. Here we show that MTM1 is expressed in blood platelets and that hundred microliters of blood is sufficient to detect the protein by western blotting. Since the most severe cases of pathogenic mutations of MTM1 lead to loss of expression of the protein, we propose that a minimal amount of blood can allow a rapid diagnostic test of X-linked myotubular myopathy, which is currently based on histopathology of muscle biopsy and molecular genetic testing. In platelets, MTM1 is a highly active 3-phosphatase mainly associated to membranes and found on the dense granules and to a lesser extent on alpha-granules. However, deletion of MTM1 in mouse had no significant effect on platelet count and on platelet secretion and aggregation induced by thrombin or collagen stimulation. Potential compensation by other members of the myotubularin family is conceivable since MTMR2 was easily detectable by western blotting and the mRNA of several members of the family increased during in vitro differentiation of human megakaryocytes and MEG-01 cells. In conclusion, we show the presence of several myotubularins in platelets and propose that minimal amounts of blood can be used to develop a rapid diagnostic test for genetic pathologies linked to loss of expression of these phosphatases.


Asunto(s)
Plaquetas/patología , Miopatías Estructurales Congénitas/diagnóstico , Proteínas Tirosina Fosfatasas no Receptoras/análisis , Animales , Plaquetas/citología , Plaquetas/metabolismo , Línea Celular , Regulación de la Expresión Génica , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Miopatías Estructurales Congénitas/sangre , Miopatías Estructurales Congénitas/genética , Agregación Plaquetaria , Proteínas Tirosina Fosfatasas no Receptoras/sangre , Proteínas Tirosina Fosfatasas no Receptoras/genética , ARN Mensajero/genética
16.
Blood ; 120(6): 1309-16, 2012 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-22677127

RESUMEN

Heparin-induced thrombocytopenia (HIT) is due primarily to IgG antibodies specific to platelet factor 4/heparin complexes (PF4/Hs) that activate platelets via FcγRIIA. CD148 is a protein tyrosine phosphatase that regulates Src kinases and collagen-induced platelet activation. Three polymorphisms affecting CD148 (Q276P, R326Q, and D872E) were studied in HIT patients and 2 control groups, with or without antibodies to PF4/Hs. Heterozygote status for CD148 276P or 326Q alleles was less frequent in HIT patients, suggesting a protective effect of these polymorphisms. Aggregation tests performed with collagen, HIT plasma, and monoclonal antibodies cross-linking FcγRIIA showed consistent hyporesponsiveness of platelets expressing the 276P/326Q alleles. In addition, platelets expressing the 276P/326Q alleles exhibited a greater sensitivity to the Src family kinases inhibitor dasatinib in response to collagen or ALB6 cross-linking FcγRIIA receptors. Moreover, the activatory phosphorylation of Src family kinases was considerably delayed as well as the phosphorylation of Linker for activation of T cells and phospholipase Cγ2, 2 major signaling proteins downstream from FcγRIIA. In conclusion, this study shows that CD148 polymorphisms affect platelet activation and probably exert a protective effect on the risk of HIT in patients with antibodies to PF4/Hs.


Asunto(s)
Heparina/efectos adversos , Activación Plaquetaria/genética , Polimorfismo de Nucleótido Simple , Receptores de IgG/fisiología , Trombocitopenia/genética , Estudios de Casos y Controles , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Heparina/inmunología , Heparina/metabolismo , Humanos , Masculino , Factor Plaquetario 4/inmunología , Factor Plaquetario 4/metabolismo , Polimorfismo de Nucleótido Simple/fisiología , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/fisiología , Receptores de IgG/genética , Receptores de IgG/metabolismo , Factores de Riesgo , Trombocitopenia/sangre , Trombocitopenia/etiología
17.
Blood ; 120(8): 1703-12, 2012 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-22776819

RESUMEN

Although estrogens are known to have a deleterious effect on the venous thrombosis risk and a preventive action on the development of arterial atheroma, their effect on platelet function in vivo remains unclear. Here, we demonstrate that a chronic high physiologic level of estradiol (E2) in mice leads to a marked decrease in platelet responsiveness ex vivo and in vivo compared with ovariectomized controls. E2 treatment led to increased bleeding time and a resistance to thromboembolism. Hematopoietic chimera mice harboring a selective deletion of estrogen receptors (ERs) α or ß were used to demonstrate that the effects of E2 were exclusively because of hematopoietic ERα. Within ERα the activation function-1 domain was not required for resistance to thromboembolism, as was previously shown for atheroprotection. This domain is mandatory for E2-mediated reproductive function and suggests that this role is controlled independently. Differential proteomics indicated that E2 treatment modulated the expression of platelet proteins including ß1 tubulin and a few other proteins that may impact platelet production and activation. Overall, these data demonstrate a previously unrecognized role for E2 in regulating the platelet proteome and platelet function, and point to new potential antithrombotic and vasculoprotective therapeutic strategies.


Asunto(s)
Plaquetas/efectos de los fármacos , Estradiol/uso terapéutico , Receptor alfa de Estrógeno/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Tromboembolia/prevención & control , Animales , Tiempo de Sangría , Plaquetas/citología , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Femenino , Eliminación de Gen , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ovariectomía , Proteoma/metabolismo , Tromboembolia/genética , Tromboembolia/metabolismo , Tubulina (Proteína)/metabolismo
18.
Adv Biol Regul ; 91: 100992, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37793962

RESUMEN

Blood platelets are produced by megakaryocytes through a complex program of differentiation and play a critical role in hemostasis and thrombosis. These anucleate cells are the target of antithrombotic drugs that prevent them from clumping in cardiovascular disease conditions. Platelets also significantly contribute to various aspects of physiopathology, including interorgan communications, healing, inflammation, and thromboinflammation. Their production and activation are strictly regulated by highly elaborated mechanisms. Among them, those involving inositol lipids have drawn the attention of researchers. Phosphoinositides represent the seven combinatorially phosphorylated forms of the inositol head group of inositol lipids. They play a crucial role in regulating intracellular mechanisms, such as signal transduction, actin cytoskeleton rearrangements, and membrane trafficking, either by generating second messengers or by directly binding to specific domains of effector proteins. In this review, we will explore how phosphoinositides are implicated in controlling platelet production by megakaryocytes and in platelet activation processes. We will also discuss the diversity of phosphoinositides in platelets, their role in granule biogenesis and maintenance, as well as in integrin signaling. Finally, we will address the discovery of a novel pool of phosphatidylinositol 3-monophosphate in the outerleaflet of the plasma membrane of human and mouse platelets.


Asunto(s)
Plaquetas , Trombosis , Animales , Ratones , Humanos , Plaquetas/patología , Fosfatidilinositoles/metabolismo , Inflamación , Trombosis/metabolismo , Inositol/metabolismo
19.
JHEP Rep ; 6(1): 100930, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38149074

RESUMEN

Background & Aims: The constitutive androstane receptor (CAR) is a nuclear receptor that binds diverse xenobiotics and whose activation leads to the modulation of the expression of target genes involved in xenobiotic detoxification and energy metabolism. Although CAR hepatic activity is considered to be higher in women than in men, its sex-dependent response to an acute pharmacological activation has seldom been investigated. Methods: The hepatic transcriptome, plasma markers, and hepatic metabolome, were analysed in Car+/+ and Car-/- male and female mice treated either with the CAR-specific agonist 1,4-bis[2-(3,5-dichloropyridyloxy)]benzene (TCPOBOP) or with vehicle. Results: Although 90% of TCPOBOP-sensitive genes were modulated in a sex-independent manner, the remaining 10% showed almost exclusive female liver specificity. These female-specific CAR-sensitive genes were mainly involved in xenobiotic metabolism, inflammation, and extracellular matrix organisation. CAR activation also induced higher hepatic oxidative stress and hepatocyte cytolysis in females than in males. Hepatic expression of flavin monooxygenase 3 (Fmo3) was almost abolished and was associated with a decrease in hepatic trimethylamine-N-oxide (TMAO) concentration in TCPOBOP-treated females. In line with a potential role in the control of TMAO homeostasis, CAR activation decreased platelet hyper-responsiveness in female mice supplemented with dietary choline. Conclusions: More than 10% of CAR-sensitive genes are sex-specific and influence hepatic and systemic responses such as platelet aggregation. CAR activation may be an important mechanism of sexually-dimorphic drug-induced liver injury. Impact and implications: CAR is activated by many drugs and pollutants. Its pharmacological activation had a stronger impact on hepatic gene expression and metabolism in females than in males, and had a specific impact on liver toxicity and trimethylamine metabolism. Sexual dimorphism should be considered when testing and/or prescribing xenobiotics known to activate CAR.

20.
Biochem J ; 447(1): 17-23, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22830526

RESUMEN

PtdIns3P is recognized as an important player in the control of the endocytotic pathway and in autophagy. Recent data also suggest that PtdIns3P contributes to molecular mechanisms taking place at the plasma membrane and at the midbody during cytokinesis. This lipid is present in low amounts in mammalian cells and remains difficult to quantify either by traditional techniques based on radiolabelling followed by HPLC to separate the different phosphatidylinositol monophosphates, or by high-sensitive liquid chromatography coupled to MS, which is still under development. In the present study, we describe a mass assay to quantify this lipid from various biological samples using the recombinant PtdIns3P 5-kinase, PIKfyve. Using this assay, we show an increase in the mass level of PtdIns3P in mouse and human platelets following stimulation, loss of this lipid in Vps34-deficient yeasts and its relative enrichment in early endosomes isolated from BHK cells.


Asunto(s)
Fosfatos de Fosfatidilinositol/análisis , Animales , Plaquetas/metabolismo , Cromatografía Liquida/métodos , Fosfatidilinositol 3-Quinasas Clase III/genética , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Cricetinae , Endosomas/metabolismo , Humanos , Espectrometría de Masas/métodos , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/sangre , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA