Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Platelets ; 31(6): 717-723, 2020 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-32522064

RESUMEN

Mechanisms related to platelet release in the context of the bone marrow niche are not completely known. In this review we discuss what has been discovered about four critical aspects of this process: 1) the bone marrow niche organization, 2) the role of the extracellular matrix components, 3) the mechanisms by which megakaryocytes release platelets and 4) the novel approaches to mimic the bone marrow environment and produce platelets ex vivo.


Asunto(s)
Plaquetas/metabolismo , Animales , Humanos
2.
Arterioscler Thromb Vasc Biol ; 38(9): 2041-2053, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30354258

RESUMEN

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


Asunto(s)
Hemostasis , Fosfatidilinositol 3-Quinasa/fisiología , Adhesividad Plaquetaria , Agregación Plaquetaria , Trombosis/fisiopatología , Animales , Activación Enzimática , Femenino , Humanos , Masculino , Ratones , Inhibidores de las Quinasa Fosfoinosítidos-3 , Glicoproteínas de Membrana Plaquetaria/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factor de von Willebrand/metabolismo
3.
Blood ; 125(5): 881-8, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25398937

RESUMEN

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


Asunto(s)
Plaquetas/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/genética , Glucógeno Sintasa Quinasa 3/genética , Trombosis/genética , Animales , Plaquetas/efectos de los fármacos , Plaquetas/patología , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasa Clase I/deficiencia , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Mecanotransducción Celular , Ratones , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Pirimidinonas/farmacología , Estrés Mecánico , Trombosis/enzimología , Trombosis/patología , ortoaminobenzoatos/farmacología
4.
Blood ; 124(26): 3991-5, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25305202

RESUMEN

The oral Bruton's tyrosine kinase inhibitor, ibrutinib, has recently demonstrated high efficiency in patients with relapsed B-cell malignancies. Occurrence of bleeding events has been reported in a subgroup of ibrutinib-treated patients. We demonstrate that ibrutinib selectively inhibits platelet signaling and functions downstream of the collagen receptor glycoprotein VI and strongly affects firm platelet adhesion on von Willebrand factor (VWF) under arterial flow. A longitudinal study of 14 patients indicated a correlation between occurrence of bleeding events and decreased platelet aggregation in response to collagen in platelet-rich plasma and firm adhesion on VWF under arterial flow. The addition of 50% untreated platelets was sufficient to efficiently reverse the effects of ibrutinib, and platelet functions recovered after treatment interruption as physiological platelet renewal occurred. These data have important clinical implications and provide a basis for hemostasis management during ibrutinib treatment.


Asunto(s)
Plaquetas/citología , Colágeno/química , Pirazoles/química , Pirimidinas/química , Factor de von Willebrand/química , Adenina/análogos & derivados , Plaquetas/efectos de los fármacos , Dimetilsulfóxido/química , Inhibidores Enzimáticos/química , Hemostasis , Heparina/química , Humanos , Estudios Longitudinales , Fosfolipasa C gamma/química , Fosforilación , Piperidinas , Activación Plaquetaria , Adhesividad Plaquetaria , Resistencia al Corte
5.
Adv Sci (Weinh) ; 11(18): e2308276, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38514919

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) continuously generate platelets throughout one's life. Inherited Platelet Disorders affect ≈ 3 million individuals worldwide and are characterized by defects in platelet formation or function. A critical challenge in the identification of these diseases lies in the absence of models that facilitate the study of hematopoiesis ex vivo. Here, a silk fibroin-based bioink is developed and designed for 3D bioprinting. This bioink replicates a soft and biomimetic environment, enabling the controlled differentiation of HSPCs into platelets. The formulation consisting of silk fibroin, gelatin, and alginate is fine-tuned to obtain a viscoelastic, shear-thinning, thixotropic bioink with the remarkable ability to rapidly recover after bioprinting and provide structural integrity and mechanical stability over long-term culture. Optical transparency allowed for high-resolution imaging of platelet generation, while the incorporation of enzymatic sensors allowed quantitative analysis of glycolytic metabolism during differentiation that is represented through measurable color changes. Bioprinting patient samples revealed a decrease in metabolic activity and platelet production in Inherited Platelet Disorders. These discoveries are instrumental in establishing reference ranges for classification and automating the assessment of treatment responses. This model has far-reaching implications for application in the research of blood-related diseases, prioritizing drug development strategies, and tailoring personalized therapies.


Asunto(s)
Bioimpresión , Plaquetas , Diferenciación Celular , Fibroínas , Hematopoyesis , Impresión Tridimensional , Fibroínas/metabolismo , Fibroínas/química , Bioimpresión/métodos , Humanos , Plaquetas/metabolismo , Hematopoyesis/fisiología , Tinta , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Gelatina/química
6.
Elife ; 102021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34059198

RESUMEN

Thrombocytopenic disorders have been treated with the Thrombopoietin-receptor agonist Eltrombopag. Patients with the same apparent form of thrombocytopenia may respond differently to the treatment. We describe a miniaturized bone marrow tissue model that provides a screening bioreactor for personalized, pre-treatment response prediction to Eltrombopag for individual patients. Using silk fibroin, a 3D bone marrow niche was developed that reproduces platelet biogenesis. Hematopoietic progenitors were isolated from a small amount of peripheral blood of patients with mutations in ANKRD26 and MYH9 genes, who had previously received Eltrombopag. The ex vivo response was strongly correlated with the in vivo platelet response. Induced Pluripotent Stem Cells (iPSCs) from one patient with mutated MYH9 differentiated into functional megakaryocytes that responded to Eltrombopag. Combining patient-derived cells and iPSCs with the 3D bone marrow model technology allows having a reproducible system for studying drug mechanisms and for individualized, pre-treatment selection of effective therapy in Inherited Thrombocytopenias.


Platelets are tiny cell fragments essential for blood to clot. They are created and released into the bloodstream by megakaryocytes, giant cells that live in the bone marrow. In certain genetic diseases, such as Inherited Thrombocytopenia, the bone marrow fails to produce enough platelets: this leaves patients extremely susceptible to bruising, bleeding, and poor clotting after an injury or surgery. Certain patients with Inherited Thrombocytopenia respond well to treatments designed to boost platelet production, but others do not. Why these differences exist could be investigated by designing new test systems that recreate the form and function of bone marrow in the laboratory. However, it is challenging to build the complex and poorly understood bone marrow environment outside of the body. Here, Di Buduo et al. have developed an artificial three-dimensional miniature organ bioreactor system that recreates the key features of bone marrow. In this system, megakaryocytes were grown from patient blood samples, and hooked up to a tissue scaffold made of silk. The cells were able to grow as if they were in their normal environment, and they could shed platelets into an artificial bloodstream. After treating megakaryocytes with drugs to stimulate platelet production, Di Buduo et al. found that the number of platelets recovered from the bioreactor could accurately predict which patients would respond to these drugs in the clinic. This new test system enables researchers to predict how a patient will respond to treatment, and to tailor therapy options to each individual. This technology could also be used to test new drugs for Inherited Thrombocytopenias and other blood-related diseases; if scaled-up, it could also, one day, generate large quantities of lab-grown blood cells for transfusion.


Asunto(s)
Benzoatos/farmacología , Plaquetas/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Hidrazinas/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Megacariocitos/efectos de los fármacos , Pirazoles/farmacología , Receptores de Trombopoyetina/agonistas , Nicho de Células Madre , Trombocitopenia/tratamiento farmacológico , Trombopoyesis/efectos de los fármacos , Adulto , Anciano , Reactores Biológicos , Plaquetas/metabolismo , Técnicas de Cultivo de Célula , Células Cultivadas , Femenino , Fibroínas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Megacariocitos/metabolismo , Persona de Mediana Edad , Miniaturización , Mutación , Cadenas Pesadas de Miosina/genética , Receptores de Trombopoyetina/metabolismo , Trombocitopenia/sangre , Trombocitopenia/genética , Adulto Joven
7.
Blood Adv ; 5(23): 5150-5163, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34547769

RESUMEN

Hemostatic abnormalities and impaired platelet function have been described in patients affected by connective tissue disorders. We observed a moderate bleeding tendency in patients affected by collagen VI-related disorders and investigated the defects in platelet functionality, whose mechanisms are unknown. We demonstrated that megakaryocytes express collagen VI that is involved in the regulation of functional platelet production. By exploiting a collagen VI-null mouse model (Col6a1-/-), we found that collagen VI-null platelets display significantly increased susceptibility to activation and intracellular calcium signaling. Col6a1-/- megakaryocytes and platelets showed increased expression of stromal interaction molecule 1 (STIM1) and ORAI1, the components of store-operated calcium entry (SOCE), and activation of the mammalian target of rapamycin (mTOR) signaling pathway. In vivo mTOR inhibition by rapamycin reduced STIM1 and ORAI1 expression and calcium flows, resulting in a normalization of platelet susceptibility to activation. These defects were cell autonomous, because transplantation of lineage-negative bone marrow cells from Col6a1-/- mice into lethally irradiated wild-type animals showed the same alteration in SOCE and platelet activation seen in Col6a1-/- mice. Peripheral blood platelets of patients affected by collagen VI-related diseases, Bethlem myopathy and Ullrich congenital muscular dystrophy, displayed increased expression of STIM1 and ORAI1 and were more prone to activation. Altogether, these data demonstrate the importance of collagen VI in the production of functional platelets by megakaryocytes in mouse models and in collagen VI-related diseases.


Asunto(s)
Plaquetas , Señalización del Calcio , Animales , Plaquetas/metabolismo , Colágeno , Humanos , Megacariocitos/metabolismo , Ratones , Proteína ORAI1/genética , Proteína ORAI1/metabolismo
8.
Biomaterials ; 178: 122-133, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29920404

RESUMEN

In the bone marrow, the interaction of progenitor cells with the vasculature is fundamental for the release of blood cells into circulation. Silk fibroin, derived from Bombyx mori silkworm cocoons, is a promising protein biomaterial for bone marrow tissue engineering, because of its tunable architecture and mechanical properties, the capacity to incorporate labile compounds without loss of bioactivity and the demonstrated ability to support blood cell formation without premature activation. In this study, we fabricated a custom perfusion chamber to contain a multi-channel lyophilized silk sponge mimicking the vascular network in the bone marrow niche. The perfusion system consisted in an inlet and an outlet and 2 splitters that allowed funneling flow in each single channel of the silk sponge. Computational Fluid Dynamic analysis demonstrated that this design permitted confined flow inside the vascular channels. The silk channeled sponge supported efficient platelet release from megakaryocytes (Mks). After seeding, the Mks localized along SDF-1α functionalized vascular channels in the sponge. Perfusion of the channels allowed the recovery of functional platelets as demonstrated by increased PAC-1 binding upon thrombin stimulation. Further, increasing the number of channels in the silk sponge resulted in a proportional increase in the numbers of platelets recovered, suggesting applicability to scale-up for platelet production. In conclusion, we have developed a scalable system consisting of a multi-channeled silk sponge incorporated in a perfusion chamber that can provide useful technology for functional platelet production ex vivo.


Asunto(s)
Plaquetas/citología , Médula Ósea/irrigación sanguínea , Hidrodinámica , Seda/farmacología , Andamios del Tejido/química , Animales , Reactores Biológicos , Plaquetas/efectos de los fármacos , Bombyx , Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Humanos , Megacariocitos/citología , Megacariocitos/efectos de los fármacos , Reología , Seda/ultraestructura
9.
Thromb Haemost ; 115(6): 1138-46, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-26818901

RESUMEN

Antiphospholipid syndrome (APS) is an autoimmune disease characterised by the presence of antiphospholipid antibodies (aPL) associated with increased thrombotic risk and pregnancy morbidity. Although aPL are heterogeneous auto-antibodies, the major pathogenic target is the plasma protein ß2-glycoprotein 1. The molecular mechanisms of platelet activation by aPL remain poorly understood. Here, we explored the role of the class IA phosphoinositide 3-kinase (PI3K) α and ß isoforms in platelet activation by aPL. Compared to control IgG from healthy individuals, the IgG fraction isolated from patients with APS potentiates platelet aggregation induced by low dose of thrombin in vitro and increases platelet adhesion and thrombus growth on a collagen matrix under arterial shear rate through a mechanism involving glycoprotein Ib (GPIb) and Toll Like Receptor 2 (TLR-2). Using isoforms-selective pharmacological PI3K inhibitors and mice with megakaryocyte/platelet lineage-specific inactivation of class IA PI3K isoforms, we demonstrate a critical role of the PI3Kß and PI3Kα isoforms in platelet activation induced by aPL. Our data show that aPL potentiate platelet activation through GPIbα and TLR-2 via a mechanism involving the class IA PI3Kα and ß isoforms, which represent new potential therapeutic targets in the prevention or treatment of thrombotic events in patients with APS.


Asunto(s)
Anticuerpos Antifosfolípidos/sangre , Síndrome Antifosfolípido/sangre , Síndrome Antifosfolípido/inmunología , Fosfatidilinositol 3-Quinasa Clase Ia/sangre , Activación Plaquetaria/inmunología , Animales , Estudios de Casos y Controles , Fosfatidilinositol 3-Quinasa Clase I/sangre , Fosfatidilinositol 3-Quinasa Clase I/deficiencia , Fosfatidilinositol 3-Quinasa Clase I/genética , Inhibidores Enzimáticos/farmacología , Humanos , Inmunoglobulina G/sangre , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidores de las Quinasa Fosfoinosítidos-3 , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Trombosis/sangre , Trombosis/etiología , Trombosis/inmunología , Receptor Toll-Like 2/sangre
10.
Adv Biol Regul ; 61: 33-41, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26714793

RESUMEN

Blood platelets play a pivotal role in haemostasis and are strongly involved in arterial thrombosis, a leading cause of death worldwide. Besides their critical role in pathophysiology, platelets represent a valuable model to investigate, both in vitro and in vivo, the biological roles of different branches of the phosphoinositide metabolism, which is highly active in platelets. While the phospholipase C (PLC) pathway has a crucial role in platelet activation, it is now well established that at least one class I phosphoinositide 3-kinase (PI3K) is also mandatory for proper platelet functions. Except class II PI3Kγ, all other isoforms of PI3Ks (class I α, ß, γ, δ; class II α, ß and class III) are expressed in platelets. Class I PI3Ks have been extensively studied in different models over the past few decades and several isoforms are promising drug targets to treat cancer and immune diseases. In platelet activation, it has been shown that while class I PI3Kδ plays a minor role, class I PI3Kß has an important function particularly in thrombus growth and stability under high shear stress conditions found in stenotic arteries. This class I PI3K is a potentially interesting target for antithrombotic strategies. The role of class I PI3Kα remains ill defined in platelets. Herein, we will discuss our recent data showing the potential impact of inhibitors of this kinase on thrombus formation. The role of class II PI3Kα and ß as well as class III PI3K (Vps34) in platelet production and function is just emerging. Based on our data and those very recently published in the literature, we will discuss the impact of these three PI3K isoforms in platelet production and functions and in thrombosis.


Asunto(s)
Plaquetas/enzimología , Fosfatidilinositol 3-Quinasas/genética , Activación Plaquetaria/fisiología , Subunidades de Proteína/genética , Trombosis/genética , Animales , Plaquetas/citología , Plaquetas/efectos de los fármacos , Regulación de la Expresión Génica , Hemostasis/genética , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/clasificación , Isoenzimas/genética , Isoenzimas/metabolismo , Fosfatidilinositol 3-Quinasas/clasificación , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositoles/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/clasificación , Subunidades de Proteína/metabolismo , Transducción de Señal , Trombopoyesis/genética , Trombosis/enzimología , Trombosis/patología , Fosfolipasas de Tipo C/genética , Fosfolipasas de Tipo C/metabolismo
12.
Adv Biol Regul ; 54: 162-74, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24095650

RESUMEN

Class I phosphoinositide 3-kinases (PI3K) have been extensively studied in different models these last years and several isoforms are now promising drug targets to treat cancer and immune diseases. Blood platelets are non-nucleated cells critical for hemostasis and strongly involved in arterial thrombosis, a leading cause of death worldwide. Besides their role in hemostasis and thrombosis, platelets provide an interesting model to characterize the implication of the different isoforms of PI3K in signaling. They are specialized for regulated adhesion, particularly under high shear stress conditions found in arteries and use highly regulated signaling mechanisms to form and stabilize a thrombus. In this review we will highlight the role of class I PI3K in these processes and the pertinence of targeting them in the context of antithrombotic strategies but also the potential consequences on the bleeding risk of inhibiting the PI3K signaling in cancer therapy. The implication of upstream regulators of the most important isoforms of PI3K in platelets and their downstream effectors such as protein kinase B (PKB or Akt) and its target glycogen synthase kinase 3 (GSK3) will be discussed as well as the impact of PTEN and SHIP phosphatases as modulators of this pathway.


Asunto(s)
Plaquetas/enzimología , Glucógeno Sintasa Quinasa 3/metabolismo , Proteína Oncogénica v-akt/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Transducción de Señal , Trombosis/enzimología , Animales , Plaquetas/metabolismo , Glucógeno Sintasa Quinasa 3/genética , Humanos , Proteína Oncogénica v-akt/genética , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/genética , Monoéster Fosfórico Hidrolasas/genética , Activación Plaquetaria , Trombosis/genética , Trombosis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA