Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 43(10): 2042-2057, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37589138

RESUMEN

BACKGROUND: ADP-induced platelet activation leads to cell surface expression of several proteins, including TF (tissue factor). The role of ADP receptors in platelet TF modulation is still unknown. We aimed to assess the (1) involvement of P2Y1 and P2Y12 receptors in ADP-induced TF exposure; (2) modulation of TFpos-platelets in anti-P2Y12-treated patients with coronary artery disease. Based on the obtained results, we revisited the intracellular localization of TF in platelets. METHODS: The effects of P2Y1 or P2Y12 antagonists on ADP-induced TF expression and activity were analyzed in vitro by flow cytometry and thrombin generation assay in blood from healthy subjects, P2Y12-/-, and patients with gray platelet syndrome. Ex vivo, P2Y12 inhibition of TF expression by clopidogrel/prasugrel/ticagrelor, assessed by VASP (vasodilator-stimulated phosphoprotein) platelet reactivity index, was investigated in coronary artery disease (n=238). Inhibition of open canalicular system externalization and electron microscopy (TEM) were used for TF localization. RESULTS: In blood from healthy subjects, stimulated in vitro by ADP, the percentage of TFpos-platelets (17.3±5.5%) was significantly reduced in a concentration-dependent manner by P2Y12 inhibition only (-81.7±9.5% with 100 nM AR-C69931MX). In coronary artery disease, inhibition of P2Y12 is paralleled by reduction of ADP-induced platelet TF expression (VASP platelet reactivity index: 17.9±11%, 20.9±11.3%, 40.3±13%; TFpos-platelets: 10.5±4.8%, 9.8±5.9%, 13.6±6.3%, in prasugrel/ticagrelor/clopidogrel-treated patients, respectively). Despite this, 15% of clopidogrel good responders had a level of TFpos-platelets similar to the poor-responder group. Indeed, a stronger P2Y12 inhibition (130-fold) is required to inhibit TF than VASP. Thus, a VASP platelet reactivity index <20% (as in prasugrel/ticagrelor-treated patients) identifies patients with TFpos-platelets <20% (92% sensitivity). Finally, colchicine impaired in vitro ADP-induced TF expression but not α-granule release, suggesting that TF is open canalicular system stored as confirmed by TEM and platelet analysis of patients with gray platelet syndrome. CONCLUSIONS: Data show that TF expression is regulated by P2Y12 and not P2Y1; P2Y12 antagonists downregulate the percentage of TFpos-platelets. In clopidogrel good-responder patients, assessment of TFpos-platelets highlights those with residual platelet reactivity. TF is stored in open canalicular system, and its membrane exposure upon activation is prevented by colchicine.


Asunto(s)
Enfermedad de la Arteria Coronaria , Síndrome de Plaquetas Grises , Humanos , Plaquetas/metabolismo , Clopidogrel/farmacología , Enfermedad de la Arteria Coronaria/metabolismo , Síndrome de Plaquetas Grises/metabolismo , Agregación Plaquetaria , Inhibidores de Agregación Plaquetaria/farmacología , Inhibidores de Agregación Plaquetaria/uso terapéutico , Inhibidores de Agregación Plaquetaria/metabolismo , Pruebas de Función Plaquetaria/métodos , Clorhidrato de Prasugrel/metabolismo , Clorhidrato de Prasugrel/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y12 , Tromboplastina/metabolismo , Ticagrelor
2.
Nat Commun ; 14(1): 3728, 2023 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-37349339

RESUMEN

Loss of NBEAL2 function leads to grey platelet syndrome (GPS), a bleeding disorder characterized by macro-thrombocytopenia and α-granule-deficient platelets. A proportion of patients with GPS develop autoimmunity through an unknown mechanism, which might be related to the proteins NBEAL2 interacts with, specifically in immune cells. Here we show a comprehensive interactome of NBEAL2 in primary T cells, based on mass spectrometry identification of altogether 74 protein association partners. These include LRBA, a member of the same BEACH domain family as NBEAL2, recessive mutations of which cause autoimmunity and lymphocytic infiltration through defective CTLA-4 trafficking. Investigating the potential association between NBEAL2 and CTLA-4 signalling suggested by the mass spectrometry results, we confirm by co-immunoprecipitation that CTLA-4 and NBEAL2 interact with each other. Interestingly, NBEAL2 deficiency leads to low CTLA-4 expression in patient-derived effector T cells, while their regulatory T cells appear unaffected. Knocking-down NBEAL2 in healthy primary T cells recapitulates the low CTLA-4 expression observed in the T cells of GPS patients. Our results thus show that NBEAL2 is involved in the regulation of CTLA-4 expression in conventional T cells and provide a rationale for considering CTLA-4-immunoglobulin therapy in patients with GPS and autoimmune disease.


Asunto(s)
Síndrome de Plaquetas Grises , Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Plaquetas/metabolismo , Proteínas Sanguíneas/genética , Antígeno CTLA-4/genética , Antígeno CTLA-4/metabolismo , Síndrome de Plaquetas Grises/genética , Síndrome de Plaquetas Grises/metabolismo
3.
Blood ; 139(6): 922-935, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-34905616

RESUMEN

Platelet α-granules regulate hemostasis and myriad other physiological processes, but their biogenesis is unclear. Mutations in only 3 proteins are known to cause α-granule defects and bleeding disorders in humans. Two such proteins, VPS16B and VPS33B, form a complex mediating transport of newly synthesized α-granule proteins through megakaryocyte (MK) endosomal compartments. It is unclear how the VPS16B/VPS33B complex accomplishes this function. Here we report VPS16B/VPS33B associates physically with Syntaxin 12 (Stx12), a SNARE protein that mediates vesicle fusion at endosomes. Importantly, Stx12-deficient MKs display reduced α-granule numbers and overall levels of α-granule proteins, thus revealing Stx12 as a new component of the α-granule biogenesis machinery. VPS16B/VPS33B also binds CCDC22, a component of the CCC complex working at endosome exit sites. CCDC22 competes with Stx12 for binding to VPS16B/VPS33B, suggesting a possible hand-off mechanism. Moreover, the major CCC form expressed in MKs contains COMMD3, one of 10 COMMD proteins. Deficiency of COMMD3/CCDC22 causes reduced α-granule numbers and overall levels of α-granule proteins, establishing the COMMD3/CCC complex as a new factor in α-granule biogenesis. Furthermore, P-selectin traffics through the cell surface in a COMMD3-dependent manner and depletion of COMMD3 results in lysosomal degradation of P-selectin and PF4. Stx12 and COMMD3/CCC deficiency cause less severe phenotypes than VPS16B/VPS33B deficiency, suggesting Stx12 and COMMD3/CCC assist but are less important than VPS16B/VPS33B in α-granule biogenesis. Mechanistically, our results suggest VPS16B/VPS33B coordinates the endosomal entry and exit of α-granule proteins by linking the fusogenic machinery with a ubiquitous endosomal retrieval complex that is repurposed in MKs to make α-granules.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Plaquetas/metabolismo , Proteínas Qa-SNARE/metabolismo , Vesículas Secretoras/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Plaquetas/citología , Línea Celular , Síndrome de Plaquetas Grises/metabolismo , Humanos , Proteolisis
4.
Transfusion ; 59(1): 32-38, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30394544

RESUMEN

BACKGROUND: Beyond their role in hemostasis and thrombosis, platelets are also important mediators of inflammation by the release of hundreds of factors stored in their α-granules. Mutations in Nbeal2 cause gray platelet syndrome (GPS), characterized by the lack of platelet α-granules. This study aims to evaluate the immunological (proinflammatory) effects of platelet α-granules. STUDY DESIGN AND METHODS: We performed an experiment using Nbeal2-/- mice, the mouse model of GPS. Systemic inflammation was induced by intravenous injection of lipopolysaccharide (LPS). Inflammatory response was assessed by quantification of inflammatory soluble factors and platelet biological response modifiers. RESULTS: The lack of Nbeal2 (in Nbeal2 -/- mice, compared with controls) significantly reduced the recruitment of circulating neutrophils and monocytes. Moreover, after LPS injection, there was a significant increase in neutrophil and monocyte counts in control animals, compared with Nbeal2 -/- mice. The control of inflammation, evaluated by the production of anti-inflammatory cytokines, appeared to be greater in Nbeal2-/- mice compared with controls. Conversely, the production of certain inflammatory-soluble mediators known to characterize normal platelet secretion, such as soluble CD40 ligand (sCD40L), was decreased under experimental inflammation in Nbeal2 -/- mice. CONCLUSIONS: These results show that α-granules play a direct role in platelet-mediated inflammation balance, confirming the need to further investigate platelet-associated inflammatory pathophysiology and inflammatory adverse events related to blood transfusion.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Síndrome de Plaquetas Grises/inmunología , Lipopolisacáridos/toxicidad , Animales , Proteínas Sanguíneas/genética , Ligando de CD40/genética , Ligando de CD40/metabolismo , Modelos Animales de Enfermedad , Síndrome de Plaquetas Grises/genética , Síndrome de Plaquetas Grises/metabolismo , Inmunoensayo , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética
5.
Biosci Rep ; 38(5)2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30104399

RESUMEN

Platelets respond to vascular injury via surface receptor stimulation and signaling events to trigger aggregation, procoagulant activation, and granule secretion during hemostasis, thrombosis, and vascular remodeling. Platelets contain three major types of secretory granules including dense granules (or δ-granules, DGs), α-granules (AGs), and lysosomes. The contents of platelet granules are specific. Platelet DGs store polyphosphate and small molecules such as ADP, ATP, Ca2+, and serotonin, while AGs package most of the proteins that platelets release. The platelet DGs and AGs are regarded as being budded from the endosomes and the trans-Golgi network (TGN), respectively, and then matured from multivesicular bodies (MVBs). However, the sorting machineries between DGs and AGs are different. Inherited platelet disorders are associated with deficiency of DGs and AGs, leading to bleeding diathesis in patients with Hermansky-Pudlak syndrome (HPS), gray platelet syndrome (GPS), and arthrogryposis, renal dysfunction, and cholestasis syndrome (ARC). Here, we reviewed the current understanding about how DGs differ from AGs in structure, biogenesis, and function. In particular, we focus on the sorting machineries that are involved in the formation of these two types of granules to provide insights into their diverse biological functions.


Asunto(s)
Plaquetas/metabolismo , Gránulos Citoplasmáticos/metabolismo , Cuerpos Multivesiculares/metabolismo , Vesículas Secretoras/metabolismo , Artrogriposis/metabolismo , Artrogriposis/patología , Plaquetas/patología , Colestasis/metabolismo , Colestasis/patología , Gránulos Citoplasmáticos/genética , Endosomas/metabolismo , Síndrome de Plaquetas Grises/metabolismo , Síndrome de Plaquetas Grises/patología , Síndrome de Hermanski-Pudlak/metabolismo , Síndrome de Hermanski-Pudlak/patología , Humanos , Lisosomas/metabolismo , Insuficiencia Renal/metabolismo , Insuficiencia Renal/patología , Vesículas Secretoras/genética , Red trans-Golgi/genética
6.
Arterioscler Thromb Vasc Biol ; 38(8): 1772-1784, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29930006

RESUMEN

Objective- Nbeal2-/- mice, a model of human gray platelet syndrome, have reduced neutrophil granularity and impaired host defense against systemic Staphylococcus aureus infection. We here aimed to study the role of Nbeal2 deficiency in both leukocytes and platelets during gram-negative pneumonia and sepsis. Approach and Results- We studied the role of Nbeal2 in platelets and leukocytes during murine pneumonia and sepsis by Klebsiella pneumoniae. Apart from platelet α-granule deficiency and reduced neutrophil granularity, also monocyte granularity was reduced in Nbeal2-/- mice, whereas plasma levels of MPO (myeloperoxidase), elastase, NGAL (neutrophil gelatinase-associated lipocalin), and MMP-9 (matrix metalloproteinase 9), and leukocyte CD11b expression were increased. Nbeal2-/- leukocytes showed unaltered in vitro antibacterial response and phagocytosis capacity against Klebsiella, and unchanged reactive nitrogen species and cytokine production. Also during Klebsiella pneumonia and sepsis, Nbeal2-/- mice had similar bacterial growth in lung and distant body sites, with enhanced leukocyte migration to the bronchoalveolar space. Despite similar infection-induced inflammation, organ damage was increased in Nbeal2-/- mice, which was also seen during endotoxemia. Platelet-specific Nbeal2 deficiency did not influence leukocyte functions, indicating that Nbeal2 directly modifies leukocytes. Transfusion of Nbeal2-/- but not of Nbeal2+/+ platelets into thrombocytopenic mice was associated with bleeding in the lung but similar host defense, pointing at a role for platelet α-granules in maintaining vascular integrity but not host defense during Klebsiella pneumosepsis. Conclusions- These data show that Nbeal2 deficiency-resulting in gray platelet syndrome-affects platelets, neutrophils, and monocytes, with intact host defense but increased organ damage during gram-negative pneumosepsis.


Asunto(s)
Plaquetas/metabolismo , Proteínas Sanguíneas/deficiencia , Síndrome de Plaquetas Grises/metabolismo , Infecciones por Klebsiella/metabolismo , Klebsiella pneumoniae/patogenicidad , Insuficiencia Multiorgánica/metabolismo , Neumonía Bacteriana/metabolismo , Sepsis/metabolismo , Animales , Plaquetas/microbiología , Proteínas Sanguíneas/genética , Antígeno CD11b/sangre , Modelos Animales de Enfermedad , Femenino , Síndrome de Plaquetas Grises/sangre , Síndrome de Plaquetas Grises/genética , Interacciones Huésped-Patógeno , Infecciones por Klebsiella/sangre , Infecciones por Klebsiella/genética , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/crecimiento & desarrollo , Lipocalina 2/sangre , Masculino , Metaloproteinasa 9 de la Matriz/sangre , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Monocitos/microbiología , Insuficiencia Multiorgánica/sangre , Insuficiencia Multiorgánica/genética , Insuficiencia Multiorgánica/microbiología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Elastasa Pancreática/sangre , Peroxidasa/sangre , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Transfusión de Plaquetas , Neumonía Bacteriana/sangre , Neumonía Bacteriana/genética , Neumonía Bacteriana/microbiología , Sepsis/sangre , Sepsis/genética , Sepsis/microbiología
7.
Platelets ; 29(6): 632-635, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29869935

RESUMEN

Homozygosity/compound heterozygosity for loss of function mutations in neurobeachin-like 2 (NBEAL2) is causative for Gray platelet syndrome (GPS; MIM #139090), characterized by thrombocytopenia and large platelets lacking α-granules and cargo. Most GPS-associated NBEAL2 mutations generate nonsense codons; frameshifts causing premature translation termination and/or changes in mRNA splicing have also been observed. Data regarding NBEAL2 protein expression in GPS patients is limited. We observed absence of NBEAL2 in platelets from GPS patients with 3 different genotypes, and reduced/truncated platelet NBEAL2 has been reported for others. GPS is commonly associated with mild bleeding, but lifethreatening bleeding has been reported in some cases. A common long-term complication in GPS patients is myelofibrosis; splenomegaly is less common but sometimes of sufficient severity to merit splenectomy. Like GPS patients, mice lacking NBEAL2 expression exhibit macrothrombocytopenia, deficiency of platelet α-granules, splenomegaly, myelofibrosis, impaired platelet function and abnormalities in megakaryocyte development. Animal studies have also reported impaired platelet function in vivo using laser injury and thrombo-inflammation models. NBEAL2 is a large gene with 54 exons, and several putative functional domains have been identified in NBEAL2, including PH (pleckstrin homology) and BEACH (beige and Chediak-Higashi) domains shared with other members of a protein family that includes LYST and LRBA, also expressed by hematopoietic cells. Potential NBEAL2-interacting proteins have recently been identified, and it is expected that current and future efforts will reveal the cellular mechanisms by which NBEAL2 facilitates platelet development and supports hemostatic function.


Asunto(s)
Plaquetas/metabolismo , Proteínas Sanguíneas/genética , Síndrome de Plaquetas Grises/genética , Mutación , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Proteínas Sanguíneas/metabolismo , Niño , Preescolar , Femenino , Síndrome de Plaquetas Grises/sangre , Síndrome de Plaquetas Grises/metabolismo , Hemorragia/sangre , Hemorragia/genética , Hemorragia/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
8.
Platelets ; 28(2): 147-154, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28277061

RESUMEN

Platelets are critical to hemostasis and thrombosis. Upon detecting injury, platelets show a range of responses including the release of protein cargo from α-granules. This cargo is synthesized by platelet precursor megakaryocytes or endocytosed by megakaryocytes and/or platelets. Insights into α-granule biogenesis have come from studies of hereditary conditions where these granules are immature, deficient or absent. Studies of Arthrogryposis, Renal dysfunction, and Cholestasis (ARC) syndrome identified the first proteins essential to α-granule biogenesis: VPS33B and VPS16B. VPS33B and VPS16B form a complex, and in the absence of either, platelets lack α-granules and the granule-specific membrane protein P-selectin. Gray Platelet Syndrome (GPS) platelets also lack conventionally recognizable α-granules, although P-selectin containing structures are present. GPS arises from mutations affecting NBEAL2. The GPS phenotype is more benign than ARC syndrome, but it can cause life-threatening bleeding, progressive thrombocytopenia, and myelofibrosis. We review the essential roles of VPS33B, VPS16B, and NBEAL2 in α-granule development. We also examine the existing data on their mechanisms of action, where many details remain poorly understood. VPS33B and VPS16B are ubiquitously expressed and ARC syndrome is a multisystem disorder that causes lethality early in life. Thus, VPS33B and VPS16B are clearly involved in other processes besides α-granule biogenesis. Studies of their involvement in vesicular trafficking and protein interactions are reviewed to gain insights into their roles in α-granule formation. NBEAL2 mutations primarily affect megakaryocytes and platelets, and while little is known about NBEAL2 function some insights can be gained from studies of related proteins, such as LYST.


Asunto(s)
Plaquetas/metabolismo , Gránulos Citoplasmáticos/metabolismo , Animales , Artrogriposis/diagnóstico , Artrogriposis/etiología , Artrogriposis/metabolismo , Transporte Biológico , Plaquetas/ultraestructura , Colestasis/diagnóstico , Colestasis/etiología , Colestasis/metabolismo , Gránulos Citoplasmáticos/ultraestructura , Síndrome de Plaquetas Grises/diagnóstico , Síndrome de Plaquetas Grises/etiología , Síndrome de Plaquetas Grises/metabolismo , Humanos , Megacariocitos/metabolismo , Mutación , Fenotipo , Insuficiencia Renal/diagnóstico , Insuficiencia Renal/etiología , Insuficiencia Renal/metabolismo , Vesículas Secretoras/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
9.
Sci Rep ; 6: 23213, 2016 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-26987485

RESUMEN

The Gray Platelet Syndrome (GPS) is a rare inherited bleeding disorder characterized by deficiency of platelet α-granules, macrothrombocytopenia and marrow fibrosis. The autosomal recessive form of GPS is linked to loss of function mutations in NBEAL2, which is predicted to regulate granule trafficking in megakaryocytes, the platelet progenitors. We report the first analysis of cultured megakaryocytes from GPS patients with NBEAL2 mutations. Megakaryocytes cultured from peripheral blood or bone marrow hematopoietic progenitor cells from four patients were used to investigate megakaryopoiesis, megakaryocyte morphology and platelet formation. In vitro differentiation of megakaryocytes was normal, whereas we observed deficiency of megakaryocyte α-granule proteins and emperipolesis. Importantly, we first demonstrated that platelet formation by GPS megakaryocytes was severely affected, a defect which might be the major cause of thrombocytopenia in patients. These results demonstrate that cultured megakaryocytes from GPS patients provide a valuable model to understand the pathogenesis of GPS in humans.


Asunto(s)
Plaquetas/citología , Proteínas Sanguíneas/genética , Síndrome de Plaquetas Grises/patología , Megacariocitos/citología , Anciano , Plaquetas/metabolismo , Calcio/metabolismo , Diferenciación Celular , Células Cultivadas , Niño , Emperipolesis , Síndrome de Plaquetas Grises/genética , Síndrome de Plaquetas Grises/metabolismo , Humanos , Masculino , Megacariocitos/metabolismo , Megacariocitos/patología , Modelos Biológicos , Mutación , Adulto Joven
11.
Blood ; 126(2): 133-43, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-25947942

RESUMEN

Arthrogryposis, renal dysfunction, and cholestasis (ARC) syndrome is caused by deficiencies in the trafficking proteins VPS33B or VIPAR, and is associated with a bleeding diathesis and a marked reduction in platelet α-granules. We generated a tamoxifen-inducible mouse model of VPS33B deficiency, Vps33b(fl/fl)-ER(T2), and studied the platelet phenotype and α-granule biogenesis. Ultrastructural analysis of Vps33b(fl/fl)-ER(T2) platelets identified a marked reduction in α-granule count and the presence of small granule-like structures in agreement with the platelet phenotype observed in ARC patients. A reduction of ∼65% to 75% was observed in the α-granule proteins von Willebrand factor and P-selectin. Although platelet aggregation responses were not affected, a defect in δ-granule secretion was observed. Under arteriolar shear conditions, Vps33b(fl/fl)-ER(T2) platelets were unable to form stable aggregates, and tail-bleeding measurement revealed a bleeding diathesis. Analysis of bone marrow-derived megakaryocytes (MKs) by conventional and immuno-electron microscopy from Vps33b(fl/fl)-ER(T2) mice revealed a reduction in mature type-II multivesicular bodies (MVB II) and an accumulation of large vacuoles. Proteins that are normally stored in α-granules were underrepresented in MVB II and proplatelet extensions. These results demonstrate that abnormal protein trafficking and impairment in MVB maturation in MKs underlie the α-granule deficiency in Vps33b(fl/fl)-ER(T2) mouse and ARC patients.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Megacariocitos/metabolismo , Proteínas de Transporte Vesicular/fisiología , Animales , Artrogriposis/genética , Células Cultivadas , Colestasis/genética , Síndrome de Plaquetas Grises/genética , Síndrome de Plaquetas Grises/metabolismo , Humanos , Megacariocitos/citología , Megacariocitos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Orgánulos/metabolismo , Recuento de Plaquetas , Polimorfismo de Nucleótido Simple , Transporte de Proteínas/genética , Insuficiencia Renal/genética , Proteínas de Transporte Vesicular/genética
12.
Blood ; 126(4): 531-8, 2015 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-25964667

RESUMEN

Although the biology of platelet adhesion on subendothelial matrix after vascular injury is well characterized, how the matrix biophysical properties affect platelet physiology is unknown. Here we demonstrate that geometric orientation of the matrix itself regulates platelet α-granule secretion, a key component of platelet activation. Using protein microcontact printing, we show that platelets spread beyond the geometric constraints of fibrinogen or collagen micropatterns with <5-µm features. Interestingly, α-granule exocytosis and deposition of the α-granule contents such as fibrinogen and fibronectin were primarily observed in those areas of platelet extension beyond the matrix protein micropatterns. This enables platelets to "self-deposit" additional matrix, provide more cellular membrane to extend spreading, and reinforce platelet-platelet connections. Mechanistically, this phenomenon is mediated by actin polymerization, Rac1 activation, and αIIbß3 integrin redistribution and activation, and is attenuated in gray platelet syndrome platelets, which lack α-granules, and Wiskott-Aldrich syndrome platelets, which have cytoskeletal defects. Overall, these studies demonstrate how platelets transduce geometric cues of the underlying matrix geometry into intracellular signals to extend spreading, which endows platelets spatial flexibility when spreading onto small sites of exposed subendothelium.


Asunto(s)
Plaquetas/citología , Plaquetas/metabolismo , Exocitosis/fisiología , Síndrome de Plaquetas Grises/patología , Adhesividad Plaquetaria/fisiología , Síndrome de Wiskott-Aldrich/patología , Citoesqueleto de Actina/metabolismo , Estudios de Casos y Controles , Membrana Celular/metabolismo , Células Cultivadas , Fibrinógeno/metabolismo , Fibronectinas/metabolismo , Síndrome de Plaquetas Grises/metabolismo , Humanos , Técnicas para Inmunoenzimas , Activación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Seudópodos , Síndrome de Wiskott-Aldrich/metabolismo
13.
Platelets ; 26(8): 751-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25806575

RESUMEN

The gray platelet syndrome (GPS) is a rare congenital platelet disorder characterized by mild to moderate bleeding diathesis, macrothrombocytopenia and lack of azurophilic α-granules in platelets. Some platelet and megakaryocyte (MK) abnormalities have been described, but confirmative studies of the defects in larger patient cohorts have not been undertaken. We studied platelet function and bone marrow (BM) features in five GPS patients with NBEAL2 autosomal recessive mutations from four unrelated families. In 3/3 patients, we observed a defect in platelet responses to protease-activated receptor (PAR)1-activating peptide as the most consistent finding, either isolated or combined to defective responses to other agonists. A reduction of PAR1 receptors with normal expression of major glycoproteins on the platelet surface was also found. Thrombin-induced fibrinogen binding to platelets was severely impaired in 2/2 patients. In 4/4 patients, the BM biopsy showed fibrosis (grade 2-3) and extensive emperipolesis, with many (36-65%) MKs containing 2-4 leukocytes engulfed within the cytoplasm. Reduced immunolabeling for platelet factor 4 together with normal immunolabeling for CD63 in MKs of two patients demonstrated that GPS MKs display an alpha granule-specific defect. Increased immunolabeling for P-selectin and decreased immunolabeling for PAR1, PAR4 and c-MPL were also observed in MKs of two patients. Marked emperipolesis, specific defect of MK alpha-granule content and defect of PAR1-mediated platelet responses are present in all GPS patients that we could study in detail. These results help to further characterize the disease.


Asunto(s)
Plaquetas/metabolismo , Emperipolesis , Síndrome de Plaquetas Grises/metabolismo , Megacariocitos/metabolismo , Adenosina Trifosfato/metabolismo , Anciano , Plaquetas/patología , Proteínas Sanguíneas/genética , Médula Ósea/metabolismo , Médula Ósea/patología , Estudios de Casos y Controles , Gránulos Citoplasmáticos/metabolismo , Gránulos Citoplasmáticos/patología , Fibrosis , Expresión Génica , Síndrome de Plaquetas Grises/diagnóstico , Síndrome de Plaquetas Grises/genética , Humanos , Megacariocitos/patología , Mutación , Activación Plaquetaria , Agregación Plaquetaria , Pruebas de Función Plaquetaria , Receptor PAR-1/genética , Receptor PAR-1/metabolismo
14.
PLoS One ; 10(2): e0116665, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25688860

RESUMEN

Coagulation factor XII (fXII) is important for arterial thrombosis, but its physiological activation mechanisms are unclear. In this study, we elucidated the role of platelets and platelet-derived material in fXII activation. FXII activation was only observed upon potent platelet stimulation (with thrombin, collagen-related peptide, or calcium ionophore, but not ADP) accompanied by phosphatidylserine exposure and was localised to the platelet surface. Platelets from three patients with grey platelet syndrome did not activate fXII, which suggests that platelet-associated fXII-activating material might be released from α-granules. FXII was preferentially bound by phosphotidylserine-positive platelets and annexin V abrogated platelet-dependent fXII activation; however, artificial phosphotidylserine/phosphatidylcholine microvesicles did not support fXII activation under the conditions herein. Confocal microscopy using DAPI as a poly-phosphate marker did not reveal poly-phosphates associated with an activated platelet surface. Experimental data for fXII activation indicates an auto-inhibition mechanism (ki/ka = 180 molecules/platelet). Unlike surface-associated fXII activation, platelet secretion inhibited activated fXII (fXIIa), particularly due to a released C1-inhibitor. Platelet surface-associated fXIIa formation triggered contact pathway-dependent clotting in recalcified plasma. Computer modelling suggests that fXIIa inactivation was greatly decreased in thrombi under high blood flow due to inhibitor washout. Combined, the surface-associated fXII activation and its inhibition in solution herein may be regarded as a flow-sensitive regulator that can shift the balance between surface-associated clotting and plasma-dependent inhibition, which may explain the role of fXII at high shear and why fXII is important for thrombosis but negligible in haemostasis.


Asunto(s)
Inhibidores de Factor de Coagulación Sanguínea/metabolismo , Plaquetas/metabolismo , Membrana Celular/metabolismo , Factor XII/agonistas , Factor XII/antagonistas & inhibidores , Adulto , Anciano , Anciano de 80 o más Años , Coagulación Sanguínea/efectos de los fármacos , Inhibidores de Factor de Coagulación Sanguínea/farmacología , Micropartículas Derivadas de Células/metabolismo , Proteínas Inactivadoras del Complemento 1/metabolismo , Proteínas Inactivadoras del Complemento 1/farmacología , Activación Enzimática/efectos de los fármacos , Factor XII/química , Factor XII/metabolismo , Familia , Femenino , Síndrome de Plaquetas Grises/sangre , Síndrome de Plaquetas Grises/metabolismo , Humanos , Masculino , Modelos Biológicos , Unión Proteica , Vesículas Secretoras/metabolismo
15.
Blood ; 124(24): 3624-35, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25258341

RESUMEN

NBEAL2 encodes a multidomain scaffolding protein with a putative role in granule ontogeny in human platelets. Mutations in NBEAL2 underlie gray platelet syndrome (GPS), a rare inherited bleeding disorder characterized by a lack of α-granules within blood platelets and progressive bone marrow fibrosis. We present here a novel Nbeal2(-/-) murine model of GPS and demonstrate that the lack of α-granules is due to their loss from platelets/mature megakaryocytes (MKs), and not by initial impaired formation. We show that the lack of Nbeal2 confers a proinflammatory phenotype to the bone marrow MKs, which in combination with the loss of proteins from α-granules drives the development of bone marrow fibrosis. In addition, we demonstrate that α-granule deficiency impairs platelet function beyond their purely hemostatic role and that Nbeal2 deficiency has a protective effect against cancer metastasis.


Asunto(s)
Síndrome de Plaquetas Grises/metabolismo , Megacariocitos/metabolismo , Animales , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Modelos Animales de Enfermedad , Síndrome de Plaquetas Grises/genética , Síndrome de Plaquetas Grises/patología , Humanos , Megacariocitos/patología , Ratones , Ratones Noqueados , Mutación , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología , Vesículas Secretoras
16.
Blood ; 122(19): 3349-58, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-23861251

RESUMEN

Gray platelet syndrome (GPS) is an inherited bleeding disorder associated with macrothrombocytopenia and α-granule-deficient platelets. GPS has been linked to loss of function mutations in NEABL2 (neurobeachin-like 2), and we describe here a murine GPS model, the Nbeal2(-/-) mouse. As in GPS, Nbeal2(-/-) mice exhibit splenomegaly, macrothrombocytopenia, and a deficiency of platelet α-granules and their cargo, including von Willebrand factor (VWF), thrombospondin-1, and platelet factor 4. The platelet α-granule membrane protein P-selectin is expressed at 48% of wild-type levels and externalized upon platelet activation. The presence of P-selectin and normal levels of VPS33B and VPS16B in Nbeal2(-/-) platelets suggests that NBEAL2 acts independently of VPS33B/VPS16B at a later stage of α-granule biogenesis. Impaired Nbeal2(-/-) platelet function was shown by flow cytometry, platelet aggregometry, bleeding assays, and intravital imaging of laser-induced arterial thrombus formation. Microscopic analysis detected marked abnormalities in Nbeal2(-/-) bone marrow megakaryocytes, which when cultured showed delayed maturation, decreased survival, decreased ploidy, and developmental abnormalities, including abnormal extracellular distribution of VWF. Our results confirm that α-granule secretion plays a significant role in platelet function, and they also indicate that abnormal α-granule formation in Nbeal2(-/-) mice has deleterious effects on megakaryocyte survival, development, and platelet production.


Asunto(s)
Plaquetas/patología , Proteínas Sanguíneas/genética , Médula Ósea/patología , Gránulos Citoplasmáticos/patología , Síndrome de Plaquetas Grises/patología , Megacariocitos/patología , Animales , Plaquetas/metabolismo , Proteínas Sanguíneas/deficiencia , Médula Ósea/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Diferenciación Celular , Gránulos Citoplasmáticos/metabolismo , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Síndrome de Plaquetas Grises/genética , Síndrome de Plaquetas Grises/metabolismo , Masculino , Megacariocitos/metabolismo , Ratones , Ratones Noqueados , Selectina-P/genética , Selectina-P/metabolismo , Agregación Plaquetaria , Factor Plaquetario 4/genética , Factor Plaquetario 4/metabolismo , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Trombospondina 1/genética , Trombospondina 1/metabolismo , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo
17.
Haematologica ; 98(6): 868-74, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23100277

RESUMEN

The gray platelet syndrome is a rare inherited bleeding disorder characterized by macrothrombocytopenia and deficiency of alpha (α)-granules in platelets. The genetic defect responsible for gray platelet syndrome was recently identified in biallelic mutations in the NBEAL2 gene. We studied 11 consecutive families with inherited macrothrombocytopenia of unknown origin and α-granule deficiency. All of them underwent NBEAL2 DNA sequencing and evaluation of the platelet phenotype, including a systematic assessment of the α-granule content by immunofluorescence analysis for α-granule secretory proteins. We identified 9 novel mutations hitting the two alleles of NBEAL2 in 4 probands. They included missense, nonsense and frameshift mutations, as well as nucleotide substitutions that altered the splicing mechanisms as determined at the RNA level. All the individuals with NBEAL2 biallelic mutations showed almost complete absence of platelet α-granules. Interestingly, the 13 individuals assumed to be asymptomatic because carriers of a mutated allele had platelet macrocytosis and significant reduction of the α-granule content. However, they were not thrombocytopenic. In the remaining 7 probands, we did not identify any NBEAL2 alterations, suggesting that other genetic defect(s) are responsible for their platelet phenotype. Of note, these patients were characterized by a lower severity of the α-granule deficiency than individuals with two NBEAL2 mutated alleles. Our data extend the spectrum of mutations responsible for gray platelet syndrome and demonstrate that macrothrombocytopenia with α-granule deficiency is a genetic heterogeneous trait. In terms of practical applications, the screening of NBEAL2 is worthwhile only in patients with macrothrombocytopenia and severe reduction of the α-granules. Finally, individuals carrying one NBEAL2 mutated allele have mild laboratory abnormalities, suggesting that even haploinsufficiency has an effect on platelet phenotype.


Asunto(s)
Plaquetas/metabolismo , Proteínas Sanguíneas/genética , Genotipo , Síndrome de Plaquetas Grises/genética , Síndrome de Plaquetas Grises/metabolismo , Fenotipo , Alelos , Plaquetas/ultraestructura , Exones , Estudios de Asociación Genética , Haploinsuficiencia , Humanos , Intrones , Mutación , Linaje , Empalme del ARN
18.
Blood ; 120(1): 199-206, 2012 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-22589474

RESUMEN

There has been recent controversy as to whether platelet α-granules represent a single granule population or are composed of different subpopulations that serve discrete functions. To address this question, we evaluated the localization of vesicle-associated membrane proteins (VAMPs) in spread platelets to determine whether platelets actively sort a specific subpopulation of α-granules to the periphery during spreading. Immunofluorescence microscopy demonstrated that granules expressing VAMP-3 and VAMP-8 localized to the central granulomere of spread platelets along with the granule cargos von Willebrand factor and serotonin. In contrast, α-granules expressing VAMP-7 translocated to the periphery of spread platelets along with the granule cargos TIMP2 and VEFG. Time-lapse microscopy demonstrated that α-granules expressing VAMP-7 actively moved from the granulomere to the periphery during spreading. Platelets from a patient with gray platelet syndrome lacked α-granules and demonstrated only minimal spreading. Similarly, spreading was impaired in platelets obtained from Unc13d(Jinx) mice, which are deficient in Munc13-4 and have an exocytosis defect. These studies identify a new α-granule subtype expressing VAMP-7 that moves to the periphery during spreading, supporting the premise that α-granules are heterogeneous and demonstrating that granule exocytosis is required for platelet spreading.


Asunto(s)
Plaquetas/metabolismo , Gránulos Citoplasmáticos/metabolismo , Exocitosis/fisiología , Síndrome de Plaquetas Grises/metabolismo , Proteínas R-SNARE/metabolismo , Animales , Preescolar , Femenino , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Persona de Mediana Edad , Transporte de Proteínas/fisiología , Seudópodos/metabolismo , Proteína 3 de Membrana Asociada a Vesículas/metabolismo
19.
Blood ; 118(23): 6183-91, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21989988

RESUMEN

Understanding platelet biology has been aided by studies of mice with mutations in key megakaryocytic transcription factors. We have shown that point mutations in the GATA1 cofactor FOG1 that disrupt binding to the nucleosome remodeling and deacetylase (NuRD) complex have erythroid and megakaryocyte lineages defects. Mice that are homozygous for a FOG1 point mutation (ki/ki), which ablates FOG1-NuRD interactions, have platelets that display a gray platelet syndrome (GPS)-like macrothrombocytopenia. These platelets have few α-granules and an increased number of lysosomal-like vacuoles on electron microscopy, reminiscent of the platelet in patients with GATA1-related X-linked GPS. Here we further characterized the platelet defect in ki/ki mice. We found markedly deficient levels of P-selectin protein limited to megakaryocytes and platelets. Other α-granule proteins were expressed at normal levels and were appropriately localized to α-granule-like structures. Treatment of ki/ki platelets with thrombin failed to stimulate Akt phosphorylation, resulting in poor granule secretion and platelet aggregation. These studies show that disruption of the GATA1/FOG1/NuRD transcriptional system results in a complex, pleiotropic platelet defect beyond GPS-like macrothrombocytopenia and suggest that this transcriptional complex regulates not only megakaryopoiesis but also α-granule generation and signaling pathways required for granule secretion.


Asunto(s)
Plaquetas/fisiología , Síndrome de Plaquetas Grises/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Modelos Animales de Enfermedad , Síndrome de Plaquetas Grises/metabolismo , Megacariocitos/fisiología , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Mutantes , Selectina-P/genética , Mutación Puntual/genética , Transducción de Señal/fisiología , Trombopoyesis/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...