Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 141(22): 2713-2726, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36952639

RESUMEN

Dedicator of cytokinesis (DOCK) proteins play a central role in actin cytoskeleton regulation. This is highlighted by the DOCK2 and DOCK8 deficiencies leading to actinopathies and immune deficiencies. DOCK8 and DOCK11 activate CDC42, a Rho-guanosine triphosphate hydrolases involved in actin cytoskeleton dynamics, among many cellular functions. The role of DOCK11 in human immune disease has been long suspected but, to the best of our knowledge, has never been described to date. We studied 8 male patients, from 7 unrelated families, with hemizygous DOCK11 missense variants leading to reduced DOCK11 expression. The patients were presenting with early-onset autoimmunity, including cytopenia, systemic lupus erythematosus, skin, and digestive manifestations. Patients' platelets exhibited abnormal ultrastructural morphology and spreading as well as impaired CDC42 activity. In vitro activated T cells and B-lymphoblastoid cell lines from patients exhibited aberrant protrusions and abnormal migration speed in confined channels concomitant with altered actin polymerization during migration. Knock down of DOCK11 recapitulated these abnormal cellular phenotypes in monocytes-derived dendritic cells and primary activated T cells from healthy controls. Lastly, in line with the patients' autoimmune manifestations, we also observed abnormal regulatory T-cell (Treg) phenotype with profoundly reduced FOXP3 and IKZF2 expression. Moreover, we found reduced T-cell proliferation and impaired STAT5B phosphorylation upon interleukin-2 stimulation of the patients' lymphocytes. In conclusion, DOCK11 deficiency is a new X-linked immune-related actinopathy leading to impaired CDC42 activity and STAT5 activation, and is associated with abnormal actin cytoskeleton remodeling as well as Treg phenotype, culminating in immune dysregulation and severe early-onset autoimmunity.


Asunto(s)
Enfermedades del Sistema Inmune , Síndromes de Inmunodeficiencia , Humanos , Masculino , Citoesqueleto de Actina/metabolismo , Autoinmunidad , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Enfermedades del Sistema Inmune/metabolismo , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/genética , Linfocitos T Reguladores
2.
J Cell Mol Med ; 27(15): 2228-2238, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37415307

RESUMEN

Down syndrome is the most common chromosomal abnormality in humans. Patients with Down syndrome have hematologic disorders, including mild to moderate thrombocytopenia. In case of Down syndrome, thrombocytopenia is not associated with bleeding, and it remains poorly characterized regarding molecular mechanisms. We investigated the effects of overexpression of Dyrk1A, an important factor contributing to some major Down syndrome phenotypes, on platelet number and bleeding in mice. Mice overexpressing Dyrk1A have a decrease in platelet number by 20%. However, bleeding time was found to be reduced by 50%. The thrombocytopenia and the decreased bleeding time observed were not associated to an abnormal platelet receptors expression, to a defect of platelet activation by ADP, thrombin or convulxin, to the presence of activated platelets in the circulation or to an abnormal half-life of the platelets. To propose molecular mechanisms explaining this discrepancy, we performed a network analysis of Dyrk1A interactome and demonstrated that Dyrk1A, fibronectin and fibrinogen interact indirectly through two distinct clusters of proteins. Moreover, in mice overexpressing Dyrk1A, increased plasma fibronectin and fibrinogen levels were found, linked to an increase of the hepatic fibrinogen production. Our results indicate that overexpression of Dyrk1A in mice induces decreased bleeding consistent with increased plasma fibronectin and fibrinogen levels, revealing a new role of Dyrk1A depending on its indirect interaction with these two proteins.


Asunto(s)
Síndrome de Down , Trombocitopenia , Animales , Humanos , Ratones , Plaquetas/metabolismo , Síndrome de Down/metabolismo , Fibrinógeno/metabolismo , Fibronectinas/metabolismo , Hemorragia/metabolismo , Trombocitopenia/metabolismo , Quinasas DyrK
3.
Blood ; 136(6): 740-748, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32369559

RESUMEN

The bispecific antibody emicizumab is increasingly used for hemophilia A treatment. However, its specificity for human factors IX and X (FIX and FX) has limited its in vivo functional analysis to primate models of acquired hemophilia. Here, we describe a novel mouse model that allows emicizumab function to be examined. Briefly, FVIII-deficient mice received IV emicizumab 24 hours before tail-clip bleeding was performed. A second infusion with human FIX and FX, administered 5 minutes before bleeding, generated consistent levels of emicizumab (0.7-19 mg/dL for 0.5-10 mg/kg doses) and of both FIX and FX (85 and 101 U/dL, respectively, after dosing at 100 U/kg). Plasma from these mice display FVIII-like activity in assays (diluted activated partial thromboplastin time and thrombin generation), similar to human samples containing emicizumab. Emicizumab doses of 1.5 mg/kg and higher significantly reduced blood loss in a tail-clip-bleeding model using FVIII-deficient mice. However, reduction was incomplete compared with mice treated with human FVIII concentrate, and no difference in efficacy between doses was observed. From this model, we deducted FVIII-like activity from emicizumab that corresponded to a dose of 4.5 U of FVIII per kilogram (ie, 9.0 U/dL). Interestingly, combined with a low FVIII dose (5 U/kg), emicizumab provided enough additive activity to allow complete bleeding arrest. This model could be useful for further in vivo analysis of emicizumab.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Factor IX/administración & dosificación , Factor X/administración & dosificación , Hemofilia A/tratamiento farmacológico , Hemorragia/tratamiento farmacológico , Modelos Animales , Animales , Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Biespecíficos/inmunología , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/inmunología , Quimioterapia Combinada , Factor IX/análisis , Factor IX/inmunología , Factor VIII/administración & dosificación , Factor VIII/análisis , Factor VIII/uso terapéutico , Factor X/análisis , Factor X/inmunología , Factor XIa/farmacología , Femenino , Hemofilia A/sangre , Hemofilia A/complicaciones , Hemofilia A/inmunología , Hemorragia/etiología , Infusiones Intravenosas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tiempo de Tromboplastina Parcial , Cola (estructura animal)/lesiones , Trombina/biosíntesis
4.
Arterioscler Thromb Vasc Biol ; 41(1): e1-e17, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33232198

RESUMEN

OBJECTIVE: TSP-1 (thrombospondin 1) is one of the most expressed proteins in platelet α-granules and plays an important role in the regulation of hemostasis and thrombosis. Interaction of released TSP-1 with CD47 membrane receptor has been shown to regulate major events leading to thrombus formation, such as, platelet adhesion to vascular endothelium, nitric oxide/cGMP (cyclic guanosine monophosphate) signaling, platelet activation as well as aggregation. Therefore, targeting TSP-1:CD47 axis may represent a promising antithrombotic strategy. Approach and Results: A CD47-derived cyclic peptide was engineered, namely TAX2, that targets TSP-1 and selectively prevents TSP-1:CD47 interaction. Here, we demonstrate for the first time that TAX2 peptide strongly decreases platelet aggregation and interaction with collagen under arterial shear conditions. TAX2 also delays time for complete thrombotic occlusion in 2 mouse models of arterial thrombosis following chemical injury, while Thbs1-/- mice recapitulate TAX2 effects. Importantly, TAX2 administration is not associated with increased bleeding risk or modification of hematologic parameters. CONCLUSIONS: Overall, this study sheds light on the major contribution of TSP-1:CD47 interaction in platelet activation and thrombus formation while putting forward TAX2 as an innovative antithrombotic agent with high added-value.


Asunto(s)
Arteriopatías Oclusivas/prevención & control , Antígeno CD47/antagonistas & inhibidores , Fibrinolíticos/farmacología , Péptidos Cíclicos/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Trombosis/prevención & control , Trombospondina 1/antagonistas & inhibidores , Animales , Arteriopatías Oclusivas/sangre , Arteriopatías Oclusivas/metabolismo , Antígeno CD47/metabolismo , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibrinolíticos/toxicidad , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos Cíclicos/toxicidad , Inhibidores de Agregación Plaquetaria/toxicidad , Ratas Sprague-Dawley , Transducción de Señal , Trombosis/sangre , Trombosis/metabolismo , Trombospondina 1/genética , Trombospondina 1/metabolismo , Factores de Tiempo
5.
Int J Mol Sci ; 22(16)2021 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-34445542

RESUMEN

Endoglin (Eng) is an endothelial cell (EC) transmembrane glycoprotein involved in adhesion and angiogenesis. Eng mutations result in vessel abnormalities as observed in hereditary hemorrhagic telangiectasia of type 1. The role of Eng was investigated in endothelial functions and permeability under inflammatory conditions, focusing on the actin dynamic signaling pathway. Endothelial Colony-Forming Cells (ECFC) from human cord blood and mouse lung/aortic EC (MLEC, MAEC) from Eng+/+ and Eng+/- mice were used. ECFC silenced for Eng with Eng-siRNA and ctr-siRNA were used to test tubulogenesis and permeability +/- TNFα and +/- LIM kinase inhibitors (LIMKi). In silico modeling of TNFα-Eng interactions was carried out from PDB IDs 5HZW and 5HZV. Calcium ions (Ca2+) flux was studied by Oregon Green 488 in epifluorescence microscopy. Levels of cofilin phosphorylation and tubulin post-translational modifications were evaluated by Western blot. F-actin and actin-tubulin distribution/co-localization were evaluated in cells by confocal microscopy. Eng silencing in ECFCs resulted in a decrease of cell sprouting by 50 ± 15% (p < 0.05) and an increase in pseudo-tube width (41 ± 4.5%; p < 0.001) compared to control. Upon TNFα stimulation, ECFC Eng-siRNA displayed a significant higher permeability compared to ctr-siRNA (p < 0.01), which is associated to a higher Ca2+ mobilization (p < 0.01). Computational analysis suggested that Eng mitigated TNFα activity. F-actin polymerization was significantly increased in ECFC Eng-siRNA, MAEC+/-, and MLEC+/- compared to controls (p < 0.001, p < 0.01, and p < 0.01, respectively) as well as actin/tubulin distribution (p < 0.01). Furthermore, the inactive form of cofilin (P-cofilin at Ser3) was significantly decreased by 36.7 ± 4.8% in ECFC Eng-siRNA compared to ctr-siRNA (p < 0.001). Interestingly, LIMKi reproduced the absence of Eng on TNFα-induced ECFC-increased permeability. Our data suggest that Eng plays a critical role in the homeostasis regulation of endothelial cells under inflammatory conditions (TNFα), and loss of Eng influences ECFC-related permeability through the LIMK/cofilin/actin rearrangement-signaling pathway.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Permeabilidad de la Membrana Celular , Endoglina/metabolismo , Células Endoteliales/patología , Inflamación/patología , Quinasas Lim/metabolismo , Neovascularización Patológica/patología , Factores Despolimerizantes de la Actina/genética , Animales , Endoglina/genética , Células Endoteliales/metabolismo , Inflamación/genética , Inflamación/metabolismo , Quinasas Lim/genética , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo
6.
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
7.
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
8.
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
9.
Cell Mol Life Sci ; 73(8): 1715-39, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26646071

RESUMEN

The circulatory system is walled off by different cell types, including vascular mural cells and podocytes. The interaction and interplay between endothelial cells (ECs) and mural cells, such as vascular smooth muscle cells or pericytes, play a pivotal role in vascular biology. Endoglin is an RGD-containing counter-receptor for ß1 integrins and is highly expressed by ECs during angiogenesis. We find that the adhesion between vascular ECs and mural cells is enhanced by integrin activators and inhibited upon suppression of membrane endoglin or ß1-integrin, as well as by addition of soluble endoglin (SolEng), anti-integrin α5ß1 antibody or an RGD peptide. Analysis of different endoglin mutants, allowed the mapping of the endoglin RGD motif as involved in the adhesion process. In Eng (+/-) mice, a model for hereditary hemorrhagic telangectasia type 1, endoglin haploinsufficiency induces a pericyte-dependent increase in vascular permeability. Also, transgenic mice overexpressing SolEng, an animal model for preeclampsia, show podocyturia, suggesting that SolEng is responsible for podocytes detachment from glomerular capillaries. These results suggest a critical role for endoglin in integrin-mediated adhesion of mural cells and provide a better understanding on the mechanisms of vessel maturation in normal physiology as well as in pathologies such as preeclampsia or hereditary hemorrhagic telangiectasia.


Asunto(s)
Antígenos CD/metabolismo , Adhesión Celular/fisiología , Endotelio Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Podocitos/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Antígenos CD/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Endoglina , Femenino , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Integrina beta1/genética , Células Jurkat , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Neovascularización Patológica/metabolismo , Pericitos/metabolismo , Preeclampsia/genética , Preeclampsia/patología , Embarazo , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño , Receptores de Superficie Celular/genética , Retina/metabolismo , Telangiectasia Hemorrágica Hereditaria/genética , Telangiectasia Hemorrágica Hereditaria/patología
10.
Mol Cell Proteomics ; 14(5): 1265-74, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25713122

RESUMEN

Genome-wide association studies to identify loci responsible for platelet function and cardiovascular disease susceptibility have repeatedly identified polymorphisms linked to a gene encoding platelet endothelium aggregation receptor 1 (PEAR1), an "orphan" cell surface receptor that is activated to stabilize platelet aggregates. To investigate how PEAR1 signaling is initiated, we sought to identify its extracellular ligand by creating a protein microarray representing the secretome and receptor repertoire of the human platelet. Using an avid soluble recombinant PEAR1 protein and a systematic screening assay designed to detect extracellular interactions, we identified the high affinity immunoglobulin E (IgE) receptor subunit α (FcεR1α) as a PEAR1 ligand. FcεR1α and PEAR1 directly interacted through their membrane-proximal Ig-like and 13th epidermal growth factor domains with a relatively strong affinity (KD ∼ 30 nm). Precomplexing FcεR1α with IgE potently inhibited the FcεR1α-PEAR1 interaction, and this was relieved by the anti-IgE therapeutic omalizumab. Oligomerized FcεR1α potentiated platelet aggregation and led to PEAR1 phosphorylation, an effect that was also inhibited by IgE. These findings demonstrate how a protein microarray resource can be used to gain important insight into the function of platelet receptors and provide a mechanistic basis for the initiation of PEAR1 signaling in platelet aggregation.


Asunto(s)
Plaquetas/efectos de los fármacos , Inmunoglobulina E/química , Agregación Plaquetaria/efectos de los fármacos , Receptores de Superficie Celular/química , Receptores de IgE/química , Sitios de Unión , Plaquetas/citología , Plaquetas/metabolismo , Expresión Génica , Células HEK293 , Humanos , Inmunoglobulina E/genética , Cinética , Ligandos , Omalizumab/química , Fosforilación , Análisis por Matrices de Proteínas , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Estructura Secundaria de Proteína , Receptores de Superficie Celular/genética , Receptores de IgE/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
11.
Blood ; 121(26): 5208-17, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23667054

RESUMEN

Platelet endothelial aggregation receptor-1 (PEAR1) participates in platelet aggregation via sustaining αIIbß3 activation. To investigate the role of PEAR1 in platelet formation, we monitored and manipulated PEAR1 expression in vitro in differentiating human CD34(+) hematopoietic stem cells and in vivo in zebrafish embryos. PEAR1 expression rose during CD34(+) cell differentiation up to megakaryocyte (MK) maturation. Two different lentiviral short hairpin knockdowns of PEAR1 did not affect erythropoiesis in CD34(+) cells, but increased colony-forming unit MK cell numbers twofold vs control in clonogenic assays, without substantially modifying MK maturation. The PEAR1 knockdown resulted in a twofold reduction of the phosphatase and TENsin homolog (PTEN) phosphatase expression and modulated gene expression of several phosphatidylinositol 3-kinase (PI3K)-Akt and Notch pathway genes. In zebrafish, Pear1 expression increased progressively during the first 3 days of embryo development. Both ATG and splice-blocking PEAR1 morpholinos enhanced thrombopoiesis, without affecting erythropoiesis. Western blots of 3-day-old Pear1 knockdown zebrafish revealed elevated Akt phosphorylation, coupled to transcriptional downregulation of the PTEN isoform Ptena. Neutralization by morpholinos of Ptena, but not of Ptenb, phenocopied the Pear1 zebrafish knockdown and triggered enhanced Akt phosphorylation and thrombocyte formation. In summary, this is the first demonstration that PEAR1 influences the PI3K/PTEN pathway, a critical determinant of Akt phosphorylation, itself controlling megakaryopoiesis and thrombopoiesis.


Asunto(s)
Células Madre Hematopoyéticas/citología , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Superficie Celular/metabolismo , Trombopoyesis/fisiología , Pez Cebra/crecimiento & desarrollo , Animales , Plaquetas/citología , Plaquetas/metabolismo , Western Blotting , Diferenciación Celular , Proliferación Celular , Citometría de Flujo , Células Madre Hematopoyéticas/metabolismo , Humanos , Técnicas para Inmunoenzimas , Hibridación in Situ , Megacariocitos/citología , Megacariocitos/metabolismo , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasa/genética , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Pez Cebra/metabolismo
12.
Blood ; 122(16): 2893-902, 2013 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-23945153

RESUMEN

Von Willebrand disease (VWD) type 2B is characterized by mutations causing enhanced binding of von Willebrand factor (VWF) to platelets. Bleeding tendency is associated with heterogeneous clinical manifestations, including moderate to severe thrombocytopenia. The underlying mechanism of the thrombocytopenia has remained unclear. Here, a mouse model of VWD type 2B was used to investigate pathways contributing to thrombocytopenia. Immunohistochemical analysis of blood smears revealed that mutant VWF was exclusively detected on platelets of thrombocytopenic VWD type 2B mice, suggesting that thrombocytopenic VWD type 2B mice were elevated two- to threefold upon chemical macrophage depletion. Colocalization of platelets with CD68-positive Kupffer cells and CD168-positive marginal macrophages in liver and spleen, respectively, confirmed the involvement of macrophages in the removal of VWF/platelet complexes. Significantly more platelets were found in liver and spleen of VWD type 2B mice compared with control mice. Finally, platelet survival was significantly shorter in VWD type 2B mice compared with control mice, providing a rationale for lower platelet counts in VWD type 2B mice. In conclusion, our data indicate that VWF type 2B binds to platelets and that this is a signal for clearance by macrophages, which could contribute to the thrombocytopenia in patients with VWD type 2B.


Asunto(s)
Plaquetas/citología , Macrófagos/citología , Trombocitopenia/fisiopatología , Enfermedad de von Willebrand Tipo 2/fisiopatología , Factor de von Willebrand/metabolismo , Animales , Femenino , Hemostasis , Hígado/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Perfusión , Fagocitosis , Bazo/metabolismo , Trombocitopenia/sangre , Enfermedad de von Willebrand Tipo 2/sangre
13.
Arterioscler Thromb Vasc Biol ; 34(12): 2570-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25341794

RESUMEN

OBJECTIVE: Elastin is the major structural extracellular matrix component of the arterial wall that provides the elastic recoil properties and resilience essential for proper vascular function. Elastin-derived peptides (EDP) originating from elastin fragmentation during vascular remodeling have been shown to play an important role in cell physiology and development of cardiovascular diseases. However, their involvement in thrombosis has been unexplored to date. In this study, we investigated the effects of EDP on (1) platelet aggregation and related signaling and (2) thrombus formation. We also characterized the mechanism by which EDP regulate thrombosis. APPROACH AND RESULTS: We show that EDP, derived from organo-alkaline hydrolysate of bovine insoluble elastin (kappa-elastin), decrease human platelet aggregation in whole blood induced by weak and strong agonists, such as ADP, epinephrine, arachidonic acid, collagen, TRAP, and U46619. In a mouse whole blood perfusion assay over a collagen matrix, kappa-elastin and VGVAPG, the canonical peptide recognizing the elastin receptor complex, significantly decrease thrombus formation under arterial shear conditions. We confirmed these results in vivo by demonstrating that both kappa-elastin and VGVAPG significantly prolonged the time for complete arteriole occlusion in a mouse model of thrombosis and increased tail bleeding times. Finally, we demonstrate that the regulatory role of EDP on thrombosis relies on platelets that express a functional elastin receptor complex and on the ability of EDP to disrupt plasma von Willebrand factor interaction with collagen. CONCLUSIONS: These results highlight the complex nature of the mechanisms governing thrombus formation and reveal an unsuspected regulatory role for circulating EDP in thrombosis.


Asunto(s)
Elastina/fisiología , Trombosis/etiología , Animales , Plaquetas/fisiología , Catepsina A/sangre , Bovinos , Colágeno/sangre , Elastina/sangre , Elastina/química , Humanos , Ratones , Neuraminidasa/sangre , Oligopéptidos/sangre , Oligopéptidos/química , Oligopéptidos/fisiología , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/química , Fragmentos de Péptidos/fisiología , Agregación Plaquetaria/fisiología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Proteolisis , Receptores de Superficie Celular/sangre , Transducción de Señal , Trombosis/sangre , Remodelación Vascular/fisiología , Factor de von Willebrand/metabolismo
15.
Blood ; 119(17): 4056-65, 2012 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-22371881

RESUMEN

Because single nucleotide polymorphisms (SNPs) in platelet endothelial aggregation receptor 1 (PEAR1) are associated with differential functional platelet responses in healthy subjects, we studied the function of PEAR1 in human platelets. During platelet aggregation by various agonists, the membrane expression of PEAR1 and its tyrosine phosphorylation increased. The recombinant PEAR1 EMI domain (GST-EMI) competitively reduced platelet adhesion to surface-coated PEAR1, diminished platelet aggregation, and eliminated PEAR1 phosphorylation. Polyclonal antibodies against the extracellular PEAR1 domain triggered PEAR1 phosphorylation in a src family kinase (SFK)-dependent manner. Such resulted in downstream signaling, culminating in extensive platelet degranulation and irreversible aggregation reactions interrupted by excess monovalent anti-GST-EMI F(ab) fragments. In resting platelets, the cytoplasmic tail of PEAR1 was found complexed to c-Src and Fyn, but on its phosphorylation, phospho-PEAR1 recruited p85 PI3K, resulting in persistent activation of PI3K and Akt. Thus, αIIbß3 activation was amplified, hence stabilizing platelet aggregates, a signaling cascade fully interrupted by the SFK inhibitor PP1 and the PI3K inhibitor LY294002. This study is the first demonstration of a functional role for PEAR1 in platelet activation, underpinning the observed association between PEAR1 and platelet function in genome-wide association studies.


Asunto(s)
Plaquetas/metabolismo , Agregación Plaquetaria/fisiología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Receptores de Superficie Celular/metabolismo , Familia-src Quinasas/metabolismo , Western Blotting , Comunicación Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Estudio de Asociación del Genoma Completo , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Activación Plaquetaria , Receptores de Superficie Celular/genética , Transducción de Señal , Tirosina/metabolismo
16.
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
17.
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
19.
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.

20.
J Thromb Haemost ; 21(11): 3268-3278, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37207862

RESUMEN

BACKGROUND: X-linked immunodeficiency with magnesium defect, Epstein-Barr virus infection, and neoplasia (XMEN) disease is a primary immunodeficiency due to loss-of-function mutations in the gene encoding for magnesium transporter 1 (MAGT1). Furthermore, as MAGT1 is involved in the N-glycosylation process, XMEN disease is classified as a congenital disorder of glycosylation. Although XMEN-associated immunodeficiency is well described, the mechanisms underlying platelet dysfunction and those responsible for life-threatening bleeding events have never been investigated. OBJECTIVES: To assess platelet functions in patients with XMEN disease. METHODS: Two unrelated young boys, including one before and after hematopoietic stem cell transplantation, were investigated for their platelet functions, glycoprotein expression, and serum and platelet-derived N-glycans. RESULTS: Platelet analysis highlighted abnormal elongated cells and unusual barbell-shaped proplatelets. Platelet aggregation, integrin αIIbß3 activation, calcium mobilization, and protein kinase C activity were impaired between both patients. Strikingly, platelet responses to protease-activated receptor 1 activating peptide were absent at both low and high concentrations. These defects were also associated with decreased molecular weights of glycoprotein Ibα, glycoprotein VI, and integrin αIIb due to partial impairment of N-glycosylation. All these defects were corrected after hematopoietic stem cell transplantation. CONCLUSION: Our results highlight prominent platelet dysfunction related to MAGT1 deficiency and defective N-glycosylation in several platelet proteins that could explain the hemorrhages reported in patients with XMEN disease.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Magnesio , Masculino , Humanos , Magnesio/metabolismo , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/genética , Glicosilación , Herpesvirus Humano 4/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA