Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Blood ; 134(21): 1847-1858, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31578203

RESUMEN

During thrombopoiesis, megakaryocytes (MKs) form proplatelets within the bone marrow (BM) and release platelets into BM sinusoids. Phosphoinositide-dependent protein kinase-1 (PDK1) is required for Ca2+-dependent platelet activation, but its role in MK development and regulation of platelet production remained elusive. The present study explored the role of PDK1 in the regulation of MK maturation and polarization during thrombopoiesis using a MK/platelet-specific knockout approach. Pdk1-deficient mice (Pdk1-/-) developed a significant macrothrombocytopenia as compared with wild-type mice (Pdk1fl/fl). Pdk1 deficiency further dramatically increased the number of MKs without sinusoidal contact within the BM hematopoietic compartment, resulting in a pronounced MK hyperplasia and a significantly increased extramedullary thrombopoiesis. Cultured Pdk1-/- BM-MKs showed impaired spreading on collagen, associated with an altered actin cytoskeleton structure with less filamentous actin (F-actin) and diminished podosome formation, whereas the tubulin cytoskeleton remained unaffected. This phenotype was associated with abrogated phosphorylation of p21-activated kinase (PAK) as well as its substrates LIM domain kinase and cofilin, supporting the hypothesis that the defective F-actin assembly results from increased cofilin activity in Pdk1-deficient MKs. Pdk1-/- BM-MKs developed increased ploidy and exhibited an abnormal ultrastructure with disrupted demarcation membrane system (DMS). Strikingly, Pdk1-/- BM-MKs displayed a pronounced defect in DMS polarization and produced significantly less proplatelets, indicating that PDK1 is critically required for proplatelet formation. In human MKs, genetic PDK1 knockdown resulted in increased maturity but reduced platelet-like particles formation. The present observations reveal a pivotal role of PDK1 in the regulation of MK cytoskeletal dynamics and polarization, proplatelet formation, and thrombopoiesis.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Plaquetas/metabolismo , Citoesqueleto/metabolismo , Megacariocitos/metabolismo , Trombopoyesis/fisiología , Animales , Plaquetas/citología , Humanos , Megacariocitos/citología , Ratones , Ratones Noqueados
2.
Platelets ; 32(5): 601-607, 2021 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-32726163

RESUMEN

Investigation of the bone marrow as the main compartment of hematopoiesis is critical in many research fields. Here, we adapted a centrifugation-based method for the isolation of murine bone marrow and compared it to the traditional flushing method. Analysis of primary hematopoietic stem cells, immune cells, and megakaryocytes revealed a comparable distribution of cellular (sub)populations. Furthermore, in vitro differentiated megakaryocytes displayed unaltered proplatelet formation. Strikingly, bone marrow isolation by centrifugation was considerably faster than the flushing method and significantly increased the cell yield. Thus, the centrifugation-based isolation method is highly suitable for the study of murine bone marrow cells.


Asunto(s)
Médula Ósea/metabolismo , Separación Celular/métodos , Centrifugación/métodos , Células Madre Hematopoyéticas/metabolismo , Animales , Humanos , Masculino , Ratones
3.
Blood ; 132(2): 197-209, 2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-29784641

RESUMEN

The circulating life span of blood platelets is regulated by the prosurvival protein BCL-XL It restrains the activity of BAK and BAX, the essential prodeath mediators of intrinsic apoptosis. Disabling the platelet intrinsic apoptotic pathway in mice by deleting BAK and BAX results in a doubling of platelet life span and concomitant thrombocytosis. Apoptotic platelets expose phosphatidylserine (PS) via a mechanism that is distinct from that driven by classical agonists. Whether there is any role for apoptotic PS in platelet function in vivo, however, is unclear. Apoptosis has also been associated with the platelet storage lesion (PSL), the constellation of biochemical deteriorations that occur during blood bank storage. In this study, we investigated the role of BAK/BAX-mediated apoptosis in hemostasis and thrombosis and in the development of the PSL. We show that although intrinsic apoptosis is rapidly induced during storage at 37°C, it is not detected when platelets are kept at the standard storage temperature of 22°C. Remarkably, loss of BAK and BAX did not prevent the development of the PSL at either temperature. BAK/BAX-deficient mice exhibited increased bleeding times and unstable thrombus formation. This phenotype was not caused by impaired PS exposure, but was associated with a defect in granule release from aged platelets. Strikingly, rejuvenation of BAK/BAX-deficient platelets in vivo completely rescued the observed hemostatic defects. Thus, apoptotic culling of old platelets from the bloodstream is essential to maintain a functional, hemostatically reactive platelet population. Inhibiting intrinsic apoptosis in blood banked platelets is unlikely to yield significant benefit.


Asunto(s)
Apoptosis , Plaquetas/metabolismo , Susceptibilidad a Enfermedades , Animales , Apoptosis/genética , Biomarcadores , Tiempo de Sangría , Recuento de Células Sanguíneas , Coagulación Sanguínea , Caspasas/metabolismo , Supervivencia Celular/genética , Femenino , Genotipo , Masculino , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Transducción de Señal , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
4.
Haematologica ; 105(6): 1667-1676, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31582545

RESUMEN

Platelet aggregate formation is a multistep process involving receptor-mediated, as well as biomechanical, signaling cascades, which are highly dependent on actin dynamics. We have previously shown that actin depolymerizing factor (ADF)/n-cofilin and Twinfilin 2a, members of the ADF homology (ADF-H) protein family, have distinct roles in platelet formation and function. Coactosin-like 1 (Cotl1) is another ADF-H protein that binds actin and was also shown to enhance biosynthesis of pro-inflammatory leukotrienes (LT) in granulocytes. Here, we generated mice lacking Cotl1 in the megakaryocyte lineage (Cotl1-/- ) to investigate its role in platelet production and function. Absence of Cotl1 had no impact on platelet counts, platelet activation or cytoskeletal reorganization under static conditions in vitro In contrast, Cotl1 deficiency markedly affected platelet aggregate formation on collagen and adhesion to immobilized von Willebrand factor at high shear rates in vitro, pointing to an impaired function of the platelet mechanoreceptor glycoprotein (GP) Ib. Furthermore, Cotl1 -/-platelets exhibited increased deformability at high shear rates, indicating that the GPIb defect may be linked to altered biomechanical properties of the deficient cells. In addition, we found that Cotl1 deficiency markedly affected platelet LT biosynthesis. Strikingly, exogenous LT addition restored defective aggregate formation of Cotl1-/- platelets at high shear in vitro, indicating a critical role of platelet-derived LT in thrombus formation. In vivo, Cotl1 deficiency translated into prolonged tail bleeding times and protection from occlusive arterial thrombus formation. Together, our results show that Cotl1 in platelets is an integrator of biomechanical and LT signaling in hemostasis and thrombosis.


Asunto(s)
Plaquetas , Proteínas de Microfilamentos/genética , Trombosis , Animales , Ratones , Ratones Noqueados , Activación Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria , Trombosis/genética , Factor de von Willebrand
5.
Platelets ; 31(6): 801-811, 2020 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-31948362

RESUMEN

Platelets are essential for normal hemostasis; however, pathological conditions can also trigger unwanted platelet activation precipitating thrombosis and ischemic damage of vital organs such as the heart or brain. Glycoprotein (GP)VI- and C-type lectin-like receptor 2 (CLEC-2)-mediated (hem)immunoreceptor tyrosine-based activation motif (ITAM) signaling represents a major pathway for platelet activation. The two members of the Growth-factor receptor-bound protein 2 (Grb2) family of adapter proteins expressed in platelets - Grb2 and Grb2-related adapter protein downstream of Shc (Gads) - are part of the hem(ITAM) signaling cascade by forming an adapter protein complex with linker for activation of T cells (LAT). To date, a possible functional redundancy between these two adapters in platelet activation has not been investigated. We here generated megakaryocyte- and platelet-specific Grb2/Gads double knockout (DKO) mice and analyzed their platelet function in vitro and in vivo. The DKO platelets exhibited virtually abolished (hem)ITAM signaling whereas only partial defects were seen in Grb2 or Gads single-deficient platelets. This was based on impaired phosphorylation of key molecules in the (hem)ITAM signaling cascade and translated into impaired hemostasis and partially defective arterial thrombosis, thereby exceeding the defects in either Grb2 KO or Gads KO mice. Despite this severe (hem)ITAM signaling defect, CLEC-2 dependent regulation of blood-lymphatic vessel separation was not affected in the DKO animals. These results provide direct evidence for critically redundant roles of Grb2 and Gads for platelet function in hemostasis and thrombosis, but not development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína Adaptadora GRB2/metabolismo , Motivo de Activación del Inmunorreceptor Basado en Tirosina/genética , Animales , Humanos , Ratones , Transducción de Señal
6.
Blood ; 130(25): 2774-2785, 2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-28928125

RESUMEN

Platelets, anucleated megakaryocyte (MK)-derived cells, play a major role in hemostasis and arterial thrombosis. Although protein kinase casein kinase 2 (CK2) is readily detected in MKs and platelets, the impact of CK2-dependent signaling on MK/platelet (patho-)physiology has remained elusive. The present study explored the impact of the CK2 regulatory ß-subunit on platelet biogenesis and activation. MK/platelet-specific genetic deletion of CK2ß (ck2ß-/- ) in mice resulted in a significant macrothrombocytopenia and an increased extramedullar megakaryopoiesis with an enhanced proportion of premature platelets. Although platelet life span was only mildly affected, ck2ß-/- MK displayed an abnormal microtubule structure with a drastically increased fragmentation within bone marrow and a significantly reduced proplatelet formation in vivo. In ck2ß-/- platelets, tubulin polymerization was disrupted, resulting in an impaired thrombopoiesis and an abrogated inositol 1,4,5-triphosphate receptor-dependent intracellular calcium (Ca2+) release. Presumably due to a blunted increase in the concentration of cytosolic Ca2+, activation-dependent increases of α and dense-granule secretion and integrin αIIbß3 activation, and aggregation were abrogated in ck2ß-/- platelets. Accordingly, thrombus formation and stabilization under high arterial shear rates were significantly diminished, and thrombotic vascular occlusion in vivo was significantly blunted in ck2ß-/- mice, accompanied by a slight prolongation of bleeding time. Following transient middle cerebral artery occlusion, ck2ß-/- mice displayed significantly reduced cerebral infarct volumes, developed significantly less neurological deficits, and showed significantly better outcomes after ischemic stroke than ck2ßfl/fl mice. The present observations reveal CK2ß as a novel powerful regulator of thrombopoiesis, Ca2+-dependent platelet activation, and arterial thrombosis in vivo.


Asunto(s)
Quinasa de la Caseína II/fisiología , Fragmentos de Péptidos/fisiología , Activación Plaquetaria , Trombopoyesis , Trombosis/patología , Animales , Plaquetas , Señalización del Calcio , Quinasa de la Caseína II/deficiencia , Megacariocitos/metabolismo , Megacariocitos/patología , Megacariocitos/ultraestructura , Ratones , Ratones Noqueados , Fragmentos de Péptidos/deficiencia , Trombosis/etiología , Trombosis/metabolismo
7.
Platelets ; 30(1): 9-16, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29913074

RESUMEN

Megakaryocytes differentiate from hematopoietic stem cells in the bone marrow. The transition of megakaryocytes to platelets is a complex process. Thereby, megakaryocytes extend proplatelets into sinusoidal blood vessels, where the proplatelets undergo fission to release platelets. Defects in platelet production can lead to a low platelet count (thrombocytopenia) with increased bleeding risk. Rho GTPases comprise a family of small signaling G proteins that have been shown to be master regulators of the cytoskeleton controlling many aspects of intracellular processes. The generation of Pf4-Cre transgenic mice was a major breakthrough that enabled studies in megakaryocyte-/platelet-specific knockout mouse lines and provided new insights into the central regulatory role of Rho GTPases in megakaryocyte maturation and platelet production. In this review, we will summarize major findings on the role of Rho GTPases in megakaryocyte biology with a focus on mouse lines in which knockout strategies have been applied to study the function of the best-characterized members Rac1, Cdc42 and RhoA and their downstream effector proteins.


Asunto(s)
Megacariocitos/fisiología , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo , Animales , Biomarcadores , Plaquetas/metabolismo , Citoesqueleto/metabolismo , Humanos , Trombopoyesis
8.
Platelets ; 30(6): 698-707, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30346859

RESUMEN

Inherited or acquired disorders of platelet production and function can result in thrombocytopenia and bleeding. Mouse models have proven useful for investigating the mechanisms that underlie these defects in humans. Precise methods for blood withdrawal, platelet isolation and measurement of platelet parameters are key for the generation of reproducible and conclusive data. Here, we provide three different protocols for mouse platelet isolation to encourage research knowledge transfer between experienced laboratories, while at the same time enabling less experienced researchers to implement a protocol that best suits their local expertise and equipment. We also address the issue that reported mouse platelet count and size vary considerably in the literature by investigating different factors that influence these important platelet parameters, namely: 1) genetic background and gender, 2) choice of analysis method (hematological analyzer or flow cytometry), 3) dilution of the blood sample and 4) choice of anticoagulant. The herein presented results and considerations may serve as a practical guide for both experienced and new researchers in the platelet field.


Asunto(s)
Plaquetas/metabolismo , Hematología/métodos , Recuento de Plaquetas , Animales , Masculino , Ratones
9.
Blood ; 123(22): 3381-9, 2014 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-24599546

RESUMEN

The fetal/neonatal hematopoietic system must generate enough blood cells to meet the demands of rapid growth. This unique challenge might underlie the high incidence of thrombocytopenia among preterm neonates. In this study, neonatal platelet production and turnover were investigated in newborn mice. Based on a combination of blood volume expansion and increasing platelet counts, the platelet mass increased sevenfold during the first 2 weeks of murine life, a time during which thrombopoiesis shifted from liver to bone marrow. Studies applying in vivo biotinylation and mathematical modeling showed that newborn and adult mice had similar platelet production rates, but neonatal platelets survived 1 day longer in circulation. This prolonged lifespan fully accounted for the rise in platelet counts observed during the second week of murine postnatal life. A study of pro-apoptotic and anti-apoptotic Bcl-2 family proteins showed that neonatal platelets had higher levels of the anti-apoptotic protein Bcl-2 and were more resistant to apoptosis induced by the Bcl-2/Bcl-xL inhibitor ABT-737 than adult platelets. However, genetic ablation or pharmacologic inhibition of Bcl-2 alone did not shorten neonatal platelet survival or reduce platelet counts in newborn mice, indicating the existence of redundant or alternative mechanisms mediating the prolonged lifespan of neonatal platelets.


Asunto(s)
Plaquetas/fisiología , Recuento de Plaquetas , Trombopoyesis/fisiología , Animales , Animales Recién Nacidos , Apoptosis/fisiología , Compuestos de Bifenilo/farmacología , Plaquetas/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Humanos , Recién Nacido , Hígado/citología , Volúmen Plaquetario Medio , Megacariocitos/fisiología , Megacariocitos/ultraestructura , Ratones , Nitrofenoles/farmacología , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Bazo/citología , Sulfonamidas/farmacología , Trombopoyesis/efectos de los fármacos
10.
Blood ; 122(18): 3178-87, 2013 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-23861250

RESUMEN

Blood platelets are anuclear cell fragments that are essential for blood clotting. Platelets are produced by bone marrow megakaryocytes (MKs), which extend protrusions, or so-called proplatelets, into bone marrow sinusoids. Proplatelet formation requires a profound reorganization of the MK actin and tubulin cytoskeleton. Rho GTPases, such as RhoA, Rac1, and Cdc42, are important regulators of cytoskeletal rearrangements in platelets; however, the specific roles of these proteins during platelet production have not been established. Using conditional knockout mice, we show here that Rac1 and Cdc42 possess redundant functions in platelet production and function. In contrast to a single-deficiency of either protein, a double-deficiency of Rac1 and Cdc42 in MKs resulted in macrothrombocytopenia, abnormal platelet morphology, and impaired platelet function. Double-deficient bone marrow MKs matured normally in vivo but displayed highly abnormal morphology and uncontrolled fragmentation. Consistently, a lack of Rac1/Cdc42 virtually abrogated proplatelet formation in vitro. Strikingly, this phenotype was associated with severely defective tubulin organization, whereas actin assembly and structure were barely affected. Together, these results suggest that the combined action of Rac1 and Cdc42 is crucial for platelet production, particularly by regulating microtubule dynamics.


Asunto(s)
Células Progenitoras de Megacariocitos/metabolismo , Megacariocitos/metabolismo , Tubulina (Proteína)/metabolismo , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP rac1/genética , Animales , Western Blotting , Citoesqueleto/metabolismo , Hemostasis/genética , Células Progenitoras de Megacariocitos/citología , Megacariocitos/citología , Megacariocitos/ultraestructura , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Microtúbulos/metabolismo , Seudópodos/genética , Seudópodos/metabolismo , Trombocitopenia/sangre , Trombocitopenia/genética , Trombocitopenia/metabolismo , Trombosis/sangre , Trombosis/genética , Trombosis/metabolismo , Proteína de Unión al GTP cdc42/deficiencia , Proteína de Unión al GTP rac1/deficiencia
11.
Blood ; 119(4): 1054-63, 2012 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-22045984

RESUMEN

Vascular injury initiates rapid platelet activation that is critical for hemostasis, but it also may cause thrombotic diseases, such as myocardial infarction or ischemic stroke. Reorganizations of the platelet cytoskeleton are crucial for platelet shape change and secretion and are thought to involve activation of the small GTPase RhoA. In this study, we analyzed the in vitro and in vivo consequences of megakaryocyte- and platelet-specific RhoA gene deletion in mice. We found a pronounced macrothrombocytopenia in RhoA-deficient mice, with platelet counts of approximately half that of wild-type controls. The mutant cells displayed an altered shape but only a moderately reduced life span. Shape change of RhoA-deficient platelets in response to G(13)-coupled agonists was abolished, and it was impaired in response to G(q) stimulation. Similarly, RhoA was required for efficient secretion of α and dense granules downstream of G(13) and G(q). Furthermore, RhoA was essential for integrin-mediated clot retraction but not for actomyosin rearrangements and spreading of activated platelets on fibrinogen. In vivo, RhoA deficiency resulted in markedly prolonged tail bleeding times but also significant protection in different models of arterial thrombosis and in a model of ischemic stroke. Together, these results establish RhoA as an important regulator of platelet function in thrombosis and hemostasis.


Asunto(s)
Plaquetas/patología , Hemostasis , Megacariocitos/metabolismo , Activación Plaquetaria , Trombocitopenia/fisiopatología , Trombosis/prevención & control , Proteínas de Unión al GTP rho/metabolismo , Animales , Tiempo de Sangría , Plaquetas/efectos de los fármacos , Infarto Encefálico/prevención & control , Señalización del Calcio , Forma de la Célula , Tamaño de la Célula , Retracción del Coagulo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/química , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/química , Hemostasis/efectos de los fármacos , Cinética , Megacariocitos/efectos de los fármacos , Ratones , Ratones Noqueados , Activación Plaquetaria/efectos de los fármacos , Recuento de Plaquetas , Trombocitopenia/sangre , Trombocitopenia/metabolismo , Trombocitopenia/patología , Proteínas de Unión al GTP rho/genética , Proteína de Unión al GTP rhoA
13.
Blood ; 115(16): 3364-73, 2010 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-20139097

RESUMEN

Platelet activation at sites of vascular injury is crucial for hemostasis, but it may also cause myocardial infarction or stroke. Cytoskeletal reorganization is essential for platelet activation and secretion. The small GTPase Cdc42 has been implicated as an important mediator of filopodia formation and exocytosis in various cell types, but its exact function in platelets is not established. Here, we show that the megakaryocyte/platelet-specific loss of Cdc42 leads to mild thrombocytopenia and a small increase in platelet size in mice. Unexpectedly, Cdc42-deficient platelets were able to form normally shaped filopodia and spread fully on fibrinogen upon activation, whereas filopodia formation upon selective induction of GPIb signaling was reduced compared with wild-type platelets. Furthermore, Cdc42-deficient platelets showed enhanced secretion of alpha granules, a higher adenosine diphosphate (ADP)/adenosine triphosphate (ATP) content, increased aggregation at low agonist concentrations, and enhanced aggregate formation on collagen under flow. In vivo, lack of Cdc42 resulted in faster occlusion of ferric chloride-injured arterioles. The life span of Cdc42-deficient platelets was markedly reduced, suggesting increased clearing of the cells under physiologic conditions. These data point to novel multiple functions of Cdc42 in the regulation of platelet activation, granule organization, degranulation, and a specific role in GPIb signaling.


Asunto(s)
Plaquetas/metabolismo , Plaquetas/ultraestructura , Reguladores de Proteínas de Unión al GTP/deficiencia , Hemostasis/fisiología , Activación Plaquetaria/fisiología , Animales , Western Blotting , Separación Celular , Citometría de Flujo , Ratones , Ratones Noqueados , Microscopía Electrónica
14.
Blood ; 116(10): 1767-75, 2010 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-20530287

RESUMEN

The cellular and molecular mechanisms orchestrating the complex process by which bone marrow megakaryocytes form and release platelets remain poorly understood. Mature megakaryocytes generate long cytoplasmic extensions, proplatelets, which have the capacity to generate platelets. Although microtubules are the main structural component of proplatelets and microtubule sliding is known to drive proplatelet elongation, the role of actin dynamics in the process of platelet formation has remained elusive. Here, we tailored a mouse model lacking all ADF/n-cofilin-mediated actin dynamics in megakaryocytes to specifically elucidate the role of actin filament turnover in platelet formation. We demonstrate, for the first time, that in vivo actin filament turnover plays a critical role in the late stages of platelet formation from megakaryocytes and the proper sizing of platelets in the periphery. Our results provide the genetic proof that platelet production from megakaryocytes strictly requires dynamic changes in the actin cytoskeleton.


Asunto(s)
Actinas/metabolismo , Plaquetas/metabolismo , Cofilina 1/metabolismo , Destrina/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Animales , Plaquetas/citología , Plaquetas/ultraestructura , Western Blotting , Forma de la Célula , Tamaño de la Célula , Supervivencia Celular , Cofilina 1/genética , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Destrina/genética , Fibrinógeno/metabolismo , Megacariocitos/citología , Megacariocitos/metabolismo , Megacariocitos/ultraestructura , Ratones , Ratones Noqueados , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Recuento de Plaquetas , Esplenomegalia/genética , Esplenomegalia/metabolismo , Esplenomegalia/patología , Trombina/farmacología , Factores de Tiempo
15.
Blood Adv ; 6(10): 3155-3161, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35134123

RESUMEN

G6b-B is a megakaryocyte lineage-specific immunoreceptor tyrosine-based inhibition motif-containing receptor, essential for platelet homeostasis. Mice with a genomic deletion of the entire Mpig6b locus develop severe macrothrombocytopenia and myelofibrosis, which is reflected in humans with null mutations in MPIG6B. The current model proposes that megakaryocytes lacking G6b-B develop normally, whereas proplatelet release is hampered, but the underlying molecular mechanism remains unclear. We report on a spontaneous recessive single nucleotide mutation in C57BL/6 mice, localized within the intronic region of the Mpig6b locus that abolishes G6b-B expression and reproduces macrothrombocytopenia, myelofibrosis, and osteosclerosis. As the mutation is based on a single-nucleotide exchange, Mpig6bmut mice represent an ideal model to study the role of G6b-B. Megakaryocytes from these mice were smaller, displayed a less-developed demarcation membrane system, and had a reduced expression of receptors. RNA sequencing revealed a striking global reduction in the level of megakaryocyte-specific transcripts, in conjunction with decreased protein levels of the transcription factor GATA-1 and impaired thrombopoietin signaling. The reduced number of mature MKs in the bone marrow was corroborated on a newly developed Mpig6b-null mouse strain. Our findings highlight an unexpected essential role of G6b-B in the early differentiation within the megakaryocytic lineage.


Asunto(s)
Mielofibrosis Primaria , Trombocitopenia , Animales , Plaquetas/metabolismo , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Nucleótidos/metabolismo , Mielofibrosis Primaria/genética , Trombocitopenia/genética , Trombocitopenia/metabolismo
16.
Blood Adv ; 6(17): 5184-5197, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-35819450

RESUMEN

Megakaryocytes are large cells in the bone marrow that give rise to blood platelets. Platelet biogenesis involves megakaryocyte maturation, the localization of the mature cells in close proximity to bone marrow sinusoids, and the formation of protrusions, which are elongated and shed within the circulation. Rho GTPases play important roles in platelet biogenesis and function. RhoA-deficient mice display macrothrombocytopenia and a striking mislocalization of megakaryocytes into bone marrow sinusoids and a specific defect in G-protein signaling in platelets. However, the role of the closely related protein RhoB in megakaryocytes or platelets remains unknown. In this study, we show that, in contrast to RhoA deficiency, genetic ablation of RhoB in mice results in microthrombocytopenia (decreased platelet count and size). RhoB-deficient platelets displayed mild functional defects predominantly upon induction of the collagen/glycoprotein VI pathway. Megakaryocyte maturation and localization within the bone marrow, as well as actin dynamics, were not affected in the absence of RhoB. However, in vitro-generated proplatelets revealed pronouncedly impaired microtubule organization. Furthermore, RhoB-deficient platelets and megakaryocytes displayed selective defects in microtubule dynamics/stability, correlating with reduced levels of acetylated α-tubulin. Our findings imply that the reduction of this tubulin posttranslational modification results in impaired microtubule dynamics, which might contribute to microthrombocytopenia in RhoB-deficient mice. Importantly, we demonstrate that RhoA and RhoB are localized differently and have selective, nonredundant functions in the megakaryocyte lineage.


Asunto(s)
Megacariocitos , Trombocitopenia , Proteína de Unión al GTP rhoB/metabolismo , Animales , Plaquetas/metabolismo , Megacariocitos/metabolismo , Ratones , Microtúbulos/metabolismo , Trombocitopenia/genética , Tubulina (Proteína)/metabolismo
17.
Circulation ; 121(13): 1510-7, 2010 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-20308613

RESUMEN

BACKGROUND: Blood coagulation is a tightly regulated process of sequentially activated serine proteases culminating in fibrin formation, which is critical for limiting posttraumatic blood loss but also may contribute to acute thrombotic diseases, most notably myocardial infarction and stroke. Recent studies with factor XII-deficient mice revealed that the factor XII-induced intrinsic coagulation pathway is essential for pathological thrombus formation but dispensable for hemostasis. Consequently, these findings led to the hypothesis that factor XII could be a promising pharmacological target for safe antithrombotic therapy. METHODS AND RESULTS: The complementary DNA of the previously described factor XIIa inhibitor Infestin-4, cloned from the midgut of Triatoma infestans, was fused to recombinant human albumin (rHA) and analyzed in vitro. The resulting protein rHA-Infestin-4 specifically inhibits factor XIIa and causes prolonged activated partial thromboplastin time in human, mouse, and rat plasma. To assess its inhibitory potency in vivo, mice and rats were injected with rHA-Infestin-4 and challenged in pathological thrombus formation models. In addition, bleeding assays were performed. rHA-Infestin-4 completely abolished occlusive arterial thrombus formation in mice and rats while leaving hemostasis fully intact. Furthermore, rHA-Infestin-4 was highly protective in a murine model of ischemic stroke. CONCLUSIONS: These results identify rHA-Infestin-4 as a promising agent to achieve powerful protection from ischemic cardiovascular and cerebrovascular events without affecting hemostasis.


Asunto(s)
Albúminas/farmacología , Arteriopatías Oclusivas/tratamiento farmacológico , Factor XIIa/antagonistas & inhibidores , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Proteínas de Insectos/farmacología , Trombosis/tratamiento farmacológico , Albúminas/genética , Animales , Arteriopatías Oclusivas/sangre , Isquemia Encefálica/sangre , Isquemia Encefálica/tratamiento farmacológico , ADN Complementario , Factor XIIa/genética , Factor XIIa/metabolismo , Hemostasis/efectos de los fármacos , Humanos , Infarto de la Arteria Cerebral Media/sangre , Proteínas de Insectos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Tiempo de Tromboplastina Parcial , Ratas , Ratas Endogámicas , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Trombosis/sangre
18.
Blood ; 113(9): 2056-63, 2009 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-18832659

RESUMEN

Platelet activation and aggregation at sites of vascular injury are essential for primary hemostasis, but are also major pathomechanisms underlying myocardial infarction and stroke. Changes in [Ca(2+)](i) are a central step in platelet activation. In nonexcitable cells, receptor-mediated depletion of intracellular Ca(2+) stores triggers Ca(2+) entry through store-operated calcium (SOC) channels. STIM1 has been identified as an endoplasmic reticulum (ER)-resident Ca(2+) sensor that regulates store-operated calcium entry (SOCE) in immune cells and platelets, but the identity of the platelet SOC channel has remained elusive. Orai1 (CRACM1) is the recently discovered SOC (CRAC) channel in T cells and mast cells but its role in mammalian physiology is unknown. Here we report that Orai1 is strongly expressed in human and mouse platelets. To test its role in blood clotting, we generated Orai1-deficient mice and found that their platelets display severely defective SOCE, agonist-induced Ca(2+) responses, and impaired activation and thrombus formation under flow in vitro. As a direct consequence, Orai1 deficiency in mice results in resistance to pulmonary thromboembolism, arterial thrombosis, and ischemic brain infarction, but only mild bleeding time prolongation. These results establish Orai1 as the long-sought platelet SOC channel and a crucial mediator of ischemic cardiovascular and cerebrovascular events.


Asunto(s)
Plaquetas/metabolismo , Canales de Calcio/fisiología , Calcio/metabolismo , Trombosis/genética , Animales , Plaquetas/fisiología , Peso Corporal/genética , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Canales de Calcio/genética , Canales de Calcio/metabolismo , Femenino , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Isquemia Miocárdica/genética , Isquemia Miocárdica/patología , Proteína ORAI1 , Molécula de Interacción Estromal 1 , Trombosis/metabolismo , Trombosis/patología , Trombosis/fisiopatología
19.
Blood ; 114(16): 3464-72, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19641185

RESUMEN

Damage to the integrity of the vessel wall leads to exposure of the subendothelial extracellular matrix (ECM), triggering platelet activation and aggregation. This process is essential for primary hemostasis but it may also lead to arterial thrombosis. Although the mechanisms underlying platelet activation on the ECM are well explored, it is less clear which receptors mediate cellular activation in a growing thrombus. Here we studied the role of the recently identified C-type lectin-like receptor 2 (CLEC-2) in this process. We show that anti-CLEC-2 antibody treatment of mice leads to complete and highly specific loss of CLEC-2 in circulating platelets for several days. CLEC-2-deficient platelets displayed normal adhesion under flow, but subsequent aggregate formation was severely defective in vitro and in vivo. As a consequence, CLEC-2 deficiency was associated with increased bleeding times and profound protection from occlusive arterial thrombus formation. These results reveal an essential function of CLEC-2 in hemostasis and thrombosis.


Asunto(s)
Plaquetas/metabolismo , Lectinas Tipo C/metabolismo , Adhesividad Plaquetaria , Agregación Plaquetaria , Trombosis/metabolismo , Animales , Anticuerpos/farmacología , Endotelio Vascular/metabolismo , Matriz Extracelular/metabolismo , Humanos , Lectinas Tipo C/antagonistas & inhibidores , Ratones
20.
Curr Biol ; 31(10): 2051-2064.e8, 2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-33711252

RESUMEN

Hematopoietic-specific protein 1 (Hem1) is an essential subunit of the WAVE regulatory complex (WRC) in immune cells. WRC is crucial for Arp2/3 complex activation and the protrusion of branched actin filament networks. Moreover, Hem1 loss of function in immune cells causes autoimmune diseases in humans. Here, we show that genetic removal of Hem1 in macrophages diminishes frequency and efficacy of phagocytosis as well as phagocytic cup formation in addition to defects in lamellipodial protrusion and migration. Moreover, Hem1-null macrophages displayed strong defects in cell adhesion despite unaltered podosome formation and concomitant extracellular matrix degradation. Specifically, dynamics of both adhesion and de-adhesion as well as concomitant phosphorylation of paxillin and focal adhesion kinase (FAK) were significantly compromised. Accordingly, disruption of WRC function in non-hematopoietic cells coincided with both defects in adhesion turnover and altered FAK and paxillin phosphorylation. Consistently, platelets exhibited reduced adhesion and diminished integrin αIIbß3 activation upon WRC removal. Interestingly, adhesion phenotypes, but not lamellipodia formation, were partially rescued by small molecule activation of FAK. A full rescue of the phenotype, including lamellipodia formation, required not only the presence of WRCs but also their binding to and activation by Rac. Collectively, our results uncover that WRC impacts on integrin-dependent processes in a FAK-dependent manner, controlling formation and dismantling of adhesions, relevant for properly grabbing onto extracellular surfaces and particles during cell edge expansion, like in migration or phagocytosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Adhesión Celular , Movimiento Celular , Integrinas/metabolismo , Macrófagos/metabolismo , Fagocitosis , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Quinasa 1 de Adhesión Focal/metabolismo , Masculino , Ratones , Paxillin/metabolismo , Fosforilación , Seudópodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA