Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 127(7): e166-e183, 2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32588751

RESUMEN

RATIONALE: Ca2+ signaling is a key and ubiquitous actor of cell organization and its modulation controls many cellular responses. SERCAs (sarco-endoplasmic reticulum Ca2+-ATPases) pump Ca2+ into internal stores that play a major role in the cytosolic Ca2+ concentration rise upon cell activation. Platelets exhibit 2 types of SERCAs, SERCA2b and SERCA3 (SERCA3 deficient mice), which may exert specific roles, yet ill-defined. We have recently shown that Ca2+ mobilization from SERCA3-dependent stores was required for full platelet activation in weak stimulation conditions. OBJECTIVE: To uncover the signaling mechanisms associated with Ca2+ mobilization from SERCA3-dependent stores leading to ADP secretion. METHODS AND RESULTS: Using platelets from wild-type or Serca3-deficient mice, we demonstrated that an early (within 5-10 s following stimulation) secretion of ADP specifically dependent on SERCA3 stored Ca2+ is exclusively mobilized by nicotinic acid adenosine dinucleotide-phosphate (NAADP): both Ca2+ mobilization from SERCA3-dependent stores and primary ADP secretion are blocked by the NAADP receptor antagonist Ned-19, and reciprocally both are stimulated by permeant NAADP. In contrast, Ca2+ mobilization from SERCA3-dependent stores and primary ADP secretion were unaffected by inhibition of the production of IP3 (inositol-1,4,5-trisphosphate) by phospholipase-C and accordingly were not stimulated by permeant IP3. CONCLUSIONS: Upon activation, an NAADP/SERCA3 Ca2+ mobilization pathway initiates an early ADP secretion, potentiating platelet activation, and a secondary wave of ADP secretion driven by both an IP3/SERCA2b-dependent Ca2+ stores pathway and the NAADP/SERCA3 pathway. This does not exclude that Ca2+ mobilized from SERCA3 stores may also enhance platelet global reactivity to agonists. Because of its modulating effect on platelet activation, this NAADP-SERCA3 pathway may be a relevant target for anti-thrombotic therapy. Graphic Abstract: A graphic abstract is available for this article.


Asunto(s)
Adenosina Difosfato/sangre , Comunicación Autocrina , Plaquetas/enzimología , Señalización del Calcio , NADP/análogos & derivados , Activación Plaquetaria , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/sangre , Animales , Comunicación Autocrina/efectos de los fármacos , Plaquetas/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Humanos , Inositol 1,4,5-Trifosfato/sangre , Ratones Endogámicos C57BL , Ratones Noqueados , NADP/sangre , Activación Plaquetaria/efectos de los fármacos , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Vías Secretoras , Trombina/farmacología , Tromboxano A2/sangre , Factores de Tiempo
2.
Blood ; 134(16): 1279-1288, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31471375

RESUMEN

Filamins (FLNs) are large dimeric actin-binding proteins that regulate actin cytoskeleton remodeling. In addition, FLNs serve as scaffolds for signaling proteins, such as tyrosine kinases, GTPases, or phosphatases, as well as for adhesive receptors, such as integrins. Thus, they connect adhesive receptors to signaling pathways and to cytoskeleton. There are 3 isoforms of FLN (filamin a [FLNa], FLNb, FLNc) that originate from 3 homologous genes. FLNa has been the recent focus of attention because its mutations are responsible for a wide spectrum of defects called filaminopathies A, affecting brain (peri-ventricular nodular heterotopia), heart (valve defect), skeleton, gastrointestinal tract, and, more recently, the megakaryocytic lineage. This review will focus on the physiological and pathological roles of FLNa in platelets. Indeed, FLNa mutations alter platelet production from their bone marrow precursors, the megakaryocytes, yielding giant platelets in reduced numbers (macrothrombocytopenia). In platelets per se, FLNa mutations may lead to impaired αIIbß3 integrin activation or in contrast, increased αIIbß3 activation, potentially enhancing the risk of thrombosis. Experimental work delineating the interaction of FLNa with its platelet partners, including αIIbß3, the von Willebrand factor receptor GPIb-IX-V, the tyrosine kinase Syk, and the signaling pathway of the collagen receptor GPVI, will also be reviewed.


Asunto(s)
Plaquetas/metabolismo , Filaminas/metabolismo , Animales , Humanos , Megacariocitos/metabolismo
3.
Blood ; 133(16): 1778-1788, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30602618

RESUMEN

Filamin A (FLNa) links the cell membrane with the cytoskeleton and is central in several cellular processes. Heterozygous mutations in the X-linked FLNA gene are associated with a large spectrum of conditions, including macrothrombocytopenia, called filaminopathies. Using an isogenic pluripotent stem cell model derived from patients, we show that the absence of the FLNa protein in megakaryocytes (MKs) leads to their incomplete maturation, particularly the inability to produce proplatelets. Reduction in proplatelet formation potential is associated with a defect in actomyosin contractility, which results from inappropriate RhoA activation. This dysregulated RhoA activation was observed when MKs were plated on fibrinogen but not on other matrices (fibronectin, vitronectin, collagen 1, and von Willebrand factor), strongly suggesting a role for FLNa/αIIbß3 interaction in the downregulation of RhoA activity. This was confirmed by experiments based on the overexpression of FLNa mutants deleted in the αIIbß3-binding domain and the RhoA-interacting domain, respectively. Finally, pharmacological inhibition of the RhoA-associated kinase ROCK1/2 restored a normal phenotype and proplatelet formation. Overall, this work suggests a new etiology for macrothrombocytopenia, in which increased RhoA activity is associated with disrupted FLNa/αIIbß3 interaction.


Asunto(s)
Filaminas/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Trombocitopenia/etiología , Femenino , Fibrinógeno/metabolismo , Filaminas/genética , Humanos , Megacariocitos/química , Megacariocitos/patología , Mutación , Unión Proteica/fisiología , Quinasas Asociadas a rho/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/metabolismo
4.
Blood ; 132(19): 2067-2077, 2018 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-30213874

RESUMEN

The ephrin transmembrane receptor family of tyrosine kinases is involved in platelet function. We report the first EPHB2 variant affecting platelets in 2 siblings (P1 and P2) from a consanguineous family with recurrent bleeding and normal platelet counts. Whole-exome sequencing identified a c.2233C>T variant (missense p.R745C) of the EPHB2 gene. P1 and P2 were homozygous for this variant, while their asymptomatic parents were heterozygous. The p.R745C variant within the tyrosine kinase domain was associated with defects in platelet aggregation, αIIbß3 activation, and granule secretion induced by G-protein-coupled receptor (GPCR) agonists and convulxin, as well as in thrombus formation on collagen under flow. In contrast, clot retraction, flow-dependent platelet adhesion, and spreading on fibrinogen were only mildly affected, indicating limited effects on αIIbß3 outside-in signaling. Most importantly, Lyn, Syk, and FcRγ phosphorylation, the initial steps in glycoprotein VI (GPVI) platelet signaling were drastically impaired in the absence of platelet-platelet contact, indicating a positive role for EPHB2 in GPVI activation. Likewise platelet activation by PAR4-AP showed defective Src activation, as opposed to normal protein kinase C activity and Ca2+ mobilization. Overexpression of wild-type and R745C EPHB2 variant in RBL-2H3 (rat basophilic leukemia) cells stably expressing human GPVI confirmed that EPHB2 R745C mutation impaired EPHB2 autophosphorylation but had no effect on ephrin ligand-induced EPHB2 clustering, suggesting it did not interfere with EPHB2-ephrin-mediated cell-to-cell contact. In conclusion, this novel inherited platelet disorder affecting EPHB2 demonstrates this tyrosine kinase receptor plays an important role in platelet function through crosstalk with GPVI and GPCR signaling.


Asunto(s)
Plaquetas/patología , Mutación Missense , Activación Plaquetaria , Receptor EphB2/genética , Adolescente , Plaquetas/metabolismo , Plaquetas/ultraestructura , Niño , Femenino , Humanos , Masculino , Linaje , Adhesividad Plaquetaria , Agregación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptor EphB2/metabolismo , Transducción de Señal , Adulto Joven
5.
Haematologica ; 104(12): 2493-2500, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30819911

RESUMEN

Patients with type 2B von Willebrand disease (vWD) (caused by gain-of-function mutations in the gene coding for von Willebrand factor) display bleeding to a variable extent and, in some cases, thrombocytopenia. There are several underlying causes of thrombocytopenia in type 2B vWD. It was recently suggested that desialylation-mediated platelet clearance leads to thrombocytopenia in this disease. However, this hypothesis has not been tested in vivo The relationship between platelet desialylation and the platelet count was probed in 36 patients with type 2B von Willebrand disease (p.R1306Q, p.R1341Q, and p.V1316M mutations) and in a mouse model carrying the severe p.V1316M mutation (the 2B mouse). We observed abnormally high elevated levels of platelet desialylation in both patients with the p.V1316M mutation and the 2B mice. In vitro, we demonstrated that 2B p.V1316M/von Willebrand factor induced more desialylation of normal platelets than wild-type von Willebrand factor did. Furthermore, we found that N-glycans were desialylated and we identified αIIb and ß3 as desialylation targets. Treatment of 2B mice with sialidase inhibitors (which correct platelet desialylation) was not associated with the recovery of a normal platelet count. Lastly, we demonstrated that a critical platelet desialylation threshold (not achieved in either 2B patients or 2B mice) was required to induce thrombocytopenia in vivo In conclusion, in type 2B vWD, platelet desialylation has a minor role and is not sufficient to mediate thrombocytopenia.


Asunto(s)
Plaquetas/patología , Mutación , Ácido N-Acetilneuramínico/química , Trombocitopenia/patología , Enfermedad de von Willebrand Tipo 2/complicaciones , Factor de von Willebrand/genética , Animales , Plaquetas/metabolismo , Estudios de Casos y Controles , Femenino , Estudios de Seguimiento , Humanos , Integrina alfa2beta1/metabolismo , Integrina beta3/metabolismo , Masculino , Ratones , Ácido N-Acetilneuramínico/metabolismo , Recuento de Plaquetas , Polisacáridos/metabolismo , Pronóstico , Procesamiento Proteico-Postraduccional , Trombocitopenia/etiología , Trombocitopenia/metabolismo , Enfermedad de von Willebrand Tipo 2/genética , Enfermedad de von Willebrand Tipo 2/patología
6.
Arterioscler Thromb Vasc Biol ; 38(5): 1037-1051, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29519941

RESUMEN

OBJECTIVE: Platelet secretion is crucial for many physiological platelet responses. Even though several regulators of the fusion machinery for secretory granule exocytosis have been identified in platelets, the underlying mechanisms are not yet fully characterized. APPROACH AND RESULTS: By studying a mouse model (cKO [conditional knockout]Kif5b) lacking Kif5b (kinesin-1 heavy chain) in its megakaryocytes and platelets, we evidenced unstable hemostasis characterized by an increase of blood loss associated to a marked tendency to rebleed in a tail-clip assay and thrombus instability in an in vivo thrombosis model. This instability was confirmed in vitro in a whole-blood perfusion assay under blood flow conditions. Aggregations induced by thrombin and collagen were also impaired in cKOKif5b platelets. Furthermore, P-selectin exposure, PF4 (platelet factor 4) secretion, and ATP release after thrombin stimulation were impaired in cKOKif5b platelets, highlighting the role of kinesin-1 in α-granule and dense granule secretion. Importantly, exogenous ADP rescued normal thrombin induced-aggregation in cKOKif5b platelets, which indicates that impaired aggregation was because of defective release of ADP and dense granules. Last, we demonstrated that kinesin-1 interacts with the molecular machinery comprising the granule-associated Rab27 (Ras-related protein Rab-27) protein and the Slp4 (synaptotagmin-like protein 4/SYTL4) adaptor protein. CONCLUSIONS: Our results indicate that a kinesin-1-dependent process plays a role for platelet function by acting into the mechanism underlying α-granule and dense granule secretion.


Asunto(s)
Plaquetas/enzimología , Hemostasis , Cinesinas/metabolismo , Megacariocitos/enzimología , Activación Plaquetaria , Vesículas Secretoras/enzimología , Trombosis/enzimología , Adenosina Trifosfato/sangre , Animales , Plaquetas/ultraestructura , Modelos Animales de Enfermedad , Humanos , Cinesinas/sangre , Cinesinas/deficiencia , Cinesinas/genética , Megacariocitos/ultraestructura , Ratones Endogámicos C57BL , Ratones Noqueados , Selectina-P/sangre , Agregación Plaquetaria , Factor Plaquetario 4/sangre , Vías Secretoras , Vesículas Secretoras/genética , Vesículas Secretoras/ultraestructura , Transducción de Señal , Trombosis/sangre , Trombosis/genética , Trombosis/patología , Proteínas de Transporte Vesicular/sangre , Proteínas rab27 de Unión a GTP/sangre
7.
Blood ; 128(8): 1129-38, 2016 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-27301859

RESUMEN

The role of the sarco-endoplasmic reticulum calcium (Ca(2+)) adenosine triphosphatase (ATPase) 3 (SERCA3) in platelet physiology remains poorly understood. Here, we show that SERCA3 knockout (SERCA3(-/-)) mice exhibit prolonged tail bleeding time and rebleeding. Thrombus formation was delayed both in arteries and venules in an in vivo ferric chloride-induced thrombosis model. Defective platelet adhesion and thrombus growth over collagen was confirmed in vitro. Adenosine 5'-diphosphate (ADP) removal by apyrase diminished adhesion and thrombus growth of control platelets to the level of SERCA3(-/-) platelets. Aggregation, dense granule secretion, and Ca(2+) mobilization of SERCA3(-/-) platelets induced by low collagen or low thrombin concentration were weaker than controls. Accordingly, SERCA3(-/-) platelets exhibited a partial defect in total stored Ca(2+) and in Ca(2+) store reuptake following thrombin stimulation. Importantly ADP, but not serotonin, rescued aggregation, secretion, and Ca(2+) mobilization in SERCA3(-/-) platelets, suggesting specificity. Dense granules appeared normal upon electron microscopy, mepacrine staining, and total serotonin content, ruling out a dense granule defect. ADP induced normal platelet aggregation, excluding a defect in ADP activation pathways. The SERCA3-specific inhibitor 2,5-di-(tert-butyl)-1,4-benzohydroquinone diminished both Ca(2+) mobilization and secretion of control platelets, as opposed to the SERCA2b inhibitor thapsigargin. This confirmed the specific role of catalytically active SERCA3 in ADP secretion. Accordingly, SERCA3-dependent Ca(2+) stores appeared depleted in SERCA3(-/-) platelets. Finally, αIIbß3 integrin blockade did not affect SERCA3-dependent secretion, therefore proving independent of αIIbß3 engagement. Altogether, these results show that SERCA3-dependent Ca(2+) stores control a specific ADP secretion pathway required for full platelet secretion induced by agonists at low concentration and independent of αIIbß3.


Asunto(s)
Adenosina Difosfato/metabolismo , Plaquetas/enzimología , Calcio/metabolismo , Activación Plaquetaria , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Animales , Tiempo de Sangría , Plaquetas/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Eliminación de Gen , Hemorreología/efectos de los fármacos , Hemostasis/efectos de los fármacos , Caballos , Ratones Endogámicos C57BL , Activación Plaquetaria/efectos de los fármacos , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/deficiencia , Serotonina/farmacología , Trombosis/patología
8.
Arterioscler Thromb Vasc Biol ; 37(6): 1087-1097, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28428218

RESUMEN

OBJECTIVE: Dominant mutations of the X-linked filamin A (FLNA) gene are responsible for filaminopathies A, which are rare disorders including brain periventricular nodular heterotopia, congenital intestinal pseudo-obstruction, cardiac valves or skeleton malformations, and often macrothrombocytopenia. APPROACH AND RESULTS: We studied a male patient with periventricular nodular heterotopia and congenital intestinal pseudo-obstruction, his unique X-linked FLNA allele carrying a stop codon mutation resulting in a 100-amino acid-long FLNa C-terminal extension (NP_001447.2: p.Ter2648SerextTer101). Platelet counts were normal, with few enlarged platelets. FLNa was detectable in all platelets but at 30% of control levels. Surprisingly, all platelet functions were significantly upregulated, including platelet aggregation and secretion, as induced by ADP, collagen, or von Willebrand factor in the presence of ristocetin, as well as thrombus formation in blood flow on a collagen or on a von Willebrand factor matrix. Most importantly, patient platelets stimulated with ADP exhibited a marked increase in αIIbß3 integrin activation and a parallel increase in talin recruitment to ß3, contrasting with normal Rap1 activation. These results are consistent with the mutant FLNa affecting the last step of αIIbß3 activation. Overexpression of mutant FLNa in the HEL megakaryocytic cell line correlated with an increase (compared with wild-type FLNa) in PMA-induced fibrinogen binding to and in talin and kindlin-3 recruitment by αIIbß3. CONCLUSIONS: Altogether, our results are consistent with a less binding of mutant FLNa to ß3 and the facilitated recruitment of talin by ß3 on platelet stimulation, explaining the increased αIIbß3 activation and the ensuing gain-of-platelet functions.


Asunto(s)
Plaquetas/metabolismo , Filaminas/genética , Integrina alfa2/sangre , Integrina beta3/sangre , Seudoobstrucción Intestinal/genética , Mutación , Heterotopia Nodular Periventricular/genética , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Adulto , Plaquetas/ultraestructura , Línea Celular , Análisis Mutacional de ADN , Filaminas/sangre , Predisposición Genética a la Enfermedad , Herencia , Humanos , Seudoobstrucción Intestinal/sangre , Seudoobstrucción Intestinal/diagnóstico , Masculino , Heterotopia Nodular Periventricular/sangre , Heterotopia Nodular Periventricular/diagnóstico , Fenotipo , Activación Plaquetaria , Pruebas de Función Plaquetaria , Unión Proteica , Complejo Shelterina , Transducción de Señal , Talina/sangre , Proteínas de Unión a Telómeros/sangre , Transfección , Factor de von Willebrand/metabolismo
9.
Blood ; 124(16): 2554-63, 2014 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-25061177

RESUMEN

Macrothrombocytopenias are the most important subgroup of inherited thrombocytopenias. This subgroup is particularly heterogeneous because the affected genes are involved in various functions such as cell signaling, cytoskeleton organization, and gene expression. Herein we describe the clinical and hematological features of a consanguineous family with a severe autosomal recessive macrothrombocytopenia associated with a thrombocytopathy inducing a bleeding tendency in the homozygous mutated patients. Platelet activation and cytoskeleton reorganization were impaired in these homozygous patients. Exome sequencing identified a c.222C>G mutation (missense p.74Ile>Met) in PRKACG, a gene encoding the γ-catalytic subunit of the cyclic adenosine monophosphate-dependent protein kinase, the mutated allele cosegregating with the macrothrombocytopenia. We demonstrate that the p.74Ile>Met PRKACG mutation is associated with a marked defect in proplatelet formation and a low level in filamin A in megakaryocytes (MKs). The defect in proplatelet formation was rescued in vitro by lentiviral vector-mediated overexpression of wild-type PRKACG in patient MKs. We thus conclude that PRKACG is a new central actor in platelet biogenesis and a new gene involved in inherited thrombocytopenia with giant platelets associated with a thrombocytopathy.


Asunto(s)
Plaquetas/patología , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/genética , Mutación de Línea Germinal , Megacariocitos/patología , Trombocitopenia/genética , Adulto , Plaquetas/metabolismo , Preescolar , Citoesqueleto/genética , Citoesqueleto/patología , Humanos , Lactante , Masculino , Megacariocitos/metabolismo , Linaje , Recuento de Plaquetas , Trombocitopenia/complicaciones , Trombocitopenia/patología , Adulto Joven
10.
Cell Mol Life Sci ; 72(2): 307-26, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25297919

RESUMEN

Hemostasis and pathological thrombus formation are dynamic processes that require multiple adhesive receptor-ligand interactions, with blood platelets at the heart of such events. Many studies have contributed to shed light on the importance of von Willebrand factor (VWF) interaction with its platelet receptors, glycoprotein (GP) Ib-IX-V and αIIbß3 integrin, in promoting primary platelet adhesion and aggregation following vessel injury. This review will recapitulate our current knowledge on the subject from the rheological aspect to the spatio-temporal development of thrombus formation. We will also discuss the signaling events generated by VWF/GPIb-IX-V interaction, leading to platelet activation. Additionally, we will review the growing body of evidence gathered from the recent development of pathological mouse models suggesting that VWF binding to GPIb-IX-V is a promising target in arterial and venous pathological thrombosis. Finally, the pathological aspects of VWF and its impact on platelets will be addressed.


Asunto(s)
Glicoproteínas/metabolismo , Hemostasis/fisiología , Modelos Biológicos , Adhesividad Plaquetaria/fisiología , Agregación Plaquetaria/fisiología , Trombosis/fisiopatología , Factor de von Willebrand/metabolismo , Animales , Ratones , Estructura Terciaria de Proteína , Factor de von Willebrand/genética
12.
Arterioscler Thromb Vasc Biol ; 33(1): e11-8, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23117662

RESUMEN

OBJECTIVE: We examined platelet functions in 4 unrelated patients with filaminopathy A caused by dominant mutations of the X-linked filamin A (FLNA) gene. METHODS AND RESULTS: Patients P1, P2, and P4 exhibited periventricular nodular heterotopia, heterozygozity for truncating FLNA mutations, and thrombocytopenia (except P2). P3 exhibited isolated thrombocytopenia and heterozygozity for a p.Glu1803Lys FLNA mutation. Truncated FLNA was undetectable by Western blotting of P1, P2, and P4 platelets, but full-length FLNA was detected at 37%, 82%, and 57% of control, respectively. P3 FLNA (p.Glu1803Lys and full-length) was assessed at 79%. All patients exhibited a platelet subpopulation negative for FLNA. Platelet aggregation, secretion, glycoprotein VI signaling, and thrombus growth on collagen were decreased for P1, P3, and P4, but normal for P2. For the 2 patients analyzed (P1 and P4), spreading was enhanced and, more markedly, in FLNA-negative platelets, suggesting that FLNA negatively regulates cytoskeleton reorganization. Platelet adhesion to von Willebrand factor under flow correlated with platelet full-length FLNA content: markedly reduced for P1 and P4 and unchanged for P2. Interestingly, von Willebrand factor flow adhesion was increased for P3, consistent with a gain-of-function effect enhancing glycoprotein Ib-IX-V/von Willebrand factor interaction. These results are consistent with a positive role for FLNA in platelet adhesion under high shear. CONCLUSIONS: FLNA mutation heterogeneity correlates with different platelet functional impacts and points to opposite regulatory roles of FLNA in spreading and flow adhesion under shear.


Asunto(s)
Plaquetas/metabolismo , Proteínas Contráctiles/genética , Proteínas de Microfilamentos/genética , Distrofias Musculares/sangre , Distrofias Musculares/genética , Mutación , Activación Plaquetaria/genética , Plaquetas/efectos de los fármacos , Western Blotting , Forma de la Célula/genética , Colágeno/metabolismo , Venenos de Crotálidos/farmacología , Citoesqueleto/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Fibrinógeno/metabolismo , Filaminas , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Lectinas Tipo C , Distrofias Musculares/complicaciones , Fenotipo , Activación Plaquetaria/efectos de los fármacos , Adhesividad Plaquetaria/genética , Agregación Plaquetaria/genética , Pruebas de Función Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Transducción de Señal/genética , Trombocitopenia/sangre , Trombocitopenia/genética , Trombosis/sangre , Trombosis/genética , Factor de von Willebrand/metabolismo
13.
Blood ; 118(22): 5928-37, 2011 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21960593

RESUMEN

Filaminopathies A caused by mutations in the X-linked FLNA gene are responsible for a wide spectrum of rare diseases including 2 main phenotypes, the X-linked dominant form of periventricular nodular heterotopia (FLNA-PVNH) and the otopalatodigital syndrome spectrum of disorders. In platelets, filamin A (FLNa) tethers the principal receptors ensuring the platelet-vessel wall interaction, glycoprotein Ibα and integrin αIIbß3, to the underlying cytoskeleton. Hemorrhage, coagulopathy, and thrombocytopenia are mentioned in several reports on patients with FLNA-PVNH. Abnormal platelet morphology in 2 patients with FLNA-PVNH prompted us to examine a third patient with similar platelet morphology previously diagnosed with immunologic thrombocytopenic purpura. Her enlarged platelets showed signs of FLNa degradation in Western blotting, and a heterozygous missense mutation in FLNA was detected. An irregular distribution of FLNa within the total platelet population was shown by confocal microscopy for all 3 patients. In vitro megakaryocyte cultures showed an abnormal differentiation, including an irregular distribution of FLNa with a frayed aspect, the presence of enlarged α-granules, and an abnormal fragmentation of the cytoplasm. Mutations in FLNA may represent an unrecognized cause of macrothrombocytopenia with an altered platelet production and a modified platelet-vessel wall interaction.


Asunto(s)
Proteínas Contráctiles/genética , Proteínas de Microfilamentos/genética , Mutación , Trombocitopenia/clasificación , Trombocitopenia/genética , Anciano , Células Cultivadas , Femenino , Filaminas , Predisposición Genética a la Enfermedad , Humanos , Persona de Mediana Edad , Mutación/fisiología , Recuento de Plaquetas , Síndrome , Trombocitopenia/sangre , Trombocitopenia/diagnóstico
15.
Res Pract Thromb Haemost ; 7(1): 100004, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36970741

RESUMEN

Background: Blood platelet Ca2+ stores are regulated by 2 Ca2+-ATPases (SERCA2b and SERCA3). On thrombin stimulation, nicotinic acid adenosine dinucleotide phosphate mobilizes SERCA3-dependent stores, inducing early adenosine 5'-diphosphate (ADP) secretion, potentiating later SERCA2b-dependent secretion. Objectives: The aim of this study was to identify which ADP P2 purinergic receptor (P2Y1 and/or P2Y12) is(are) involved in the amplification of platelet secretion dependent on the SERCA3-dependent Ca2+ mobilization pathway (SERCA3 stores mobilization) as triggered by low concentration of thrombin. Methods: The study used the pharmacologic antagonists MRS2719 and AR-C69931MX, of the P2Y1 and P2Y12, respectively, as well as Serca3 -/- mice and mice exhibiting platelet lineage-specific inactivation of the P2Y1 or P2Y12 genes. Results: We found that in mouse platelets, pharmacological blockade or gene inactivation of P2Y12 but not of P2Y1 led to a marked inhibition of ADP secretion after platelet stimulation with low concentration of thrombin. Likewise, in human platelets, pharmacological inhibition of P2Y12 but not of P2Y1 alters amplification of thrombin-elicited secretion through SERCA2b stores mobilization. Finally, we show that early SERCA3 stores secretion of ADP is a dense granule secretion, based on parallel adenosine triphosphate and serotonin early secretion. Furthermore, early secretion involves a single granule, based on the amount of adenosine triphosphate released. Conclusion: Altogether, these results show that at low concentrations of thrombin, SERCA3- and SERCA2b-dependent Ca2+ mobilization pathways cross-talk via ADP and activation of the P2Y12, and not the P2Y1 ADP receptor. The relevance in hemostasis of the coupling of the SERCA3 and the SERCA2b pathways is reviewed.

16.
Blood ; 115(20): 4083-92, 2010 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-20231429

RESUMEN

The role of c-Jun NH(2)-terminal kinase 1 (JNK1) in hemostasis and thrombosis remains unclear. We show here, with JNK1-deficient (JNK1(-/-)) mice, that JNK1 plays an important role in platelet biology and thrombus formation. In tail-bleeding assays, JNK1(-/-) mice exhibited longer bleeding times than wild-type mice (396 +/- 39 seconds vs 245 +/- 32 seconds). We also carried out in vitro whole-blood perfusion assays on a collagen matrix under arterial shear conditions. Thrombus formation was significantly reduced for JNK1(-/-) platelets (51%). In an in vivo model of thrombosis induced by photochemical injury to cecum vessels, occlusion times were 4.3 times longer in JNK1(-/-) arterioles than in wild-type arterioles. Moreover, in vitro studies carried out in platelet aggregation conditions demonstrated that, at low doses of agonists, platelet secretion was impaired in JNK1(-/-) platelets, leading to altered integrin alphaIIbbeta3 activation and reduced platelet aggregation, via a mechanism involving protein kinase C. JNK1 thus appears to be essential for platelet secretion in vitro, consistent with its role in thrombus growth in vivo. Finally, we showed that ERK2 and another isoform of JNK affect platelet aggregation through 2 pathways, one dependent and another independent of JNK1.


Asunto(s)
Plaquetas/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/fisiología , Agregación Plaquetaria , Trombosis/metabolismo , Animales , Coagulación Sanguínea , Western Blotting , Citometría de Flujo , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Recuento de Plaquetas , Pruebas de Función Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Proteína Quinasa C/metabolismo
17.
Blood ; 115(1): 97-106, 2010 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-19855083

RESUMEN

Protease nexin-1 (PN-1) is a serpin that inhibits plasminogen activators, plasmin, and thrombin. PN-1 is barely detectable in plasma but is expressed by platelets. Here, we studied platelet PN-1 in resting and activated conditions and its function in thrombosis. Studies on human platelets from healthy donors and from patients with a Gray platelet syndrome demonstrate that PN-1 is present both at the platelet surface and in alpha-granules. The role of PN-1 was investigated in vitro using human platelets incubated with a blocking antibody and using platelets from PN-1-deficient mice. Both approaches indicate that platelet PN-1 is active on thrombin and urokinase-type plasminogen activator. Blockade and deficiency of platelet PN-1 result in accelerated and increased tissue factor-induced thrombin generation as indicated by calibrated automated thrombography. Moreover, platelets from PN-1-deficient mice respond to subthreshold doses of thrombin, as assessed by P-selectin expression and platelet aggregation. Thrombus formation, induced ex vivo by collagen in blood flow conditions and in vivo by FeCl(3)-induced injury, is significantly increased in PN-1-deficient mice, demonstrating the antithrombotic properties of platelet PN-1. Platelet PN-1 is thus a key player in the thrombotic process, whose negative regulatory role has been, up to now, markedly underestimated.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Anticoagulantes/metabolismo , Antitrombinas/metabolismo , Plaquetas/enzimología , Receptores de Superficie Celular/metabolismo , Adulto , Animales , Circulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/enzimología , Vasos Sanguíneos/patología , Vasos Sanguíneos/fisiopatología , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Colágeno/farmacología , Glicosaminoglicanos/metabolismo , Humanos , Ratones , Agregación Plaquetaria/efectos de los fármacos , Plasma Rico en Plaquetas/metabolismo , Nexinas de Proteasas , Serpina E2 , Trombina/antagonistas & inhibidores , Tromboplastina/metabolismo , Trombosis/enzimología , Trombosis/patología , Trombosis/fisiopatología , Factores de Tiempo , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores
18.
Res Pract Thromb Haemost ; 6(2): e12672, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35316942

RESUMEN

Background: Filamin (FLN) regulates many cell functions through its scaffolding activity cross-linking cytoskeleton and integrins. FLN was shown to inhibit integrin activity, but the exact mechanism remains unclear. Objectives: The aim of this study was to evaluate the role of filamin A (FLNa) subdomains on the regulation of integrin αIIbß3 signaling. Methods: Three FLNa deletion mutants were overexpressed in the erythro-megakaryocytic leukemic cell line HEL: Del1, which lacks the N-terminal CH1-CH2 domains mediating the FLNa-actin interaction; Del2, lacking the Ig-like repeat 21, which mediates the FLNa-ß3 interaction; and Del3, lacking the C-terminal Ig repeat 24, responsible for FLNa dimerization and interaction with the small Rho guanosine triphosphatase involved in actin cytoskeleton reorganisation. Fibrinogen binding to HEL cells in suspension and talin-ß3 proximity in cells adherent to immobilized fibrinogen were assessed before and after αIIbß3 activation by the protein kinase C agonist phorbol 12-myristate 13-acetate. Results: Our results show that FLNa-actin and FLNa-ß3 interactions negatively regulate αIIbß3 activation. Moreover, FLNa-actin interaction represses Rac activation, contributing to the negative regulation of αIIbß3 activation. In contrast, the FLNa dimerization domain, which maintains Rho inactive, was found to negatively regulate αIIbß3 outside-in signaling. Conclusion: We conclude that FLNa negatively controls αIIbß3 activation by regulating actin polymerization and restraining activation of Rac, as well as outside-in signaling by repressing Rho.

19.
J Thromb Haemost ; 20(11): 2666-2678, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36006037

RESUMEN

BACKGROUND: Filaminopathies A are rare disorders affecting the brain, intestine, or skeleton, characterized by dominant X-linked filamin A (FLNA) gene mutations. Macrothrombocytopenia with functionally defective platelets is frequent. We have described a filaminopathy A male patient, exhibiting a C-terminal frame-shift FLNa mutation (Berrou et al., Arterioscler Thromb Vasc Biol. 2017;37:1087-1097). Contrasting with female patients, this male patient exhibited gain of platelet functions, including increased platelet aggregation, integrin αIIbß3 activation, and secretion at low agonist concentration, raising the issue of thrombosis risk. OBJECTIVES: Our goal is to assess the thrombotic potential of the patient FLNa mutation in an in vivo model. METHODS: We have established a mutant FlnA knock-in mouse model. RESULTS: The mutant FlnA mouse platelets phenocopied patient platelets, showing normal platelet count, lower expression level of mutant FlnA, and gain of platelet functions: increased platelet aggregation, secretion, and αIIbß3 activation, as well as increased spreading and clot retraction. Surprisingly, mutant FlnA mice exhibited a normal bleeding time, but with increased re-bleeding (77%) compared to wild type (WT) FlnA mice (27%), reflecting hemostatic plug instability. Again, in an in vivo thrombosis model, the occlusion time was not altered by the FlnA mutation, but arteriolar embolies were increased (7-fold more frequent in mutant FlnA mice versus WT mice), confirming thrombus instability. CONCLUSIONS: This study shows that the FlnA mutation found in the male patient induced gain of platelet functions in vitro, but thrombus instability in vivo. Implications for the role of FLNa in physiology of thrombus formation are discussed.


Asunto(s)
Hemostáticos , Trombosis , Masculino , Femenino , Ratones , Animales , Filaminas/genética , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Mutación con Ganancia de Función , Trombosis/genética , Trombosis/metabolismo , Mutación
20.
Exp Cell Res ; 315(5): 836-48, 2009 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-19109948

RESUMEN

In this study, we investigated the mechanism underlying Hsp27 dephosphorylation in smooth muscle cells. We found that protein phosphatase 2A (PP2A) dephosphorylates Hsp27. In addition, Hsp27 dephosphorylation was regulated by membrane cholesterol content. We showed that PDGF induced a three-fold increase in the proportion of PP2A activity regulated by cholesterol in the Triton-insoluble fraction of cell lysates. Moreover, cholesterol depletion decreased the amount of PP2A recovered in Triton-insoluble fraction. Thus, PDGF might regulate a small pool of PP2A associated with lipid rafts. Isolation of detergent-resistant membrane fragments by Optiprep-gradient density indicated that this pool of PP2A was not associated with caveolae, but was recovered in a higher density fraction (DRM-H) with ganglioside GM1, alpha-actinin, Hsp27 and p34, a component of Arp2/3 complex. These proteins were also present in dorsal ruffles containing GM1 but not caveolin-1. Phosphorylated Hsp27 levels detected in dorsal ruffles were variable. Cholesterol depletion, which inhibits dorsal ruffle formation, decreased PP2A levels and increased the Hsp27-P to Hsp27 ratio in DRM-H. These findings suggest that Hsp27 is dephosphorylated by PP2A in dorsal ruffles, in non-caveolar lipid raft microdomains. However, similarly to p34, non-phosphorylated Hsp27 is associated to non-raft membrane domains at the leading edge of lamellipodia.


Asunto(s)
Proteínas de Choque Térmico HSP27/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteína Fosfatasa 2/metabolismo , Seudópodos/metabolismo , Animales , Caveolas/enzimología , Caveolas/metabolismo , Células Cultivadas , Colesterol/farmacología , Activación Enzimática/efectos de los fármacos , Proteínas de Choque Térmico HSP27/fisiología , Microdominios de Membrana/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/ultraestructura , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Transporte de Proteínas/efectos de los fármacos , Seudópodos/efectos de los fármacos , Porcinos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA