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.
Immunity ; 45(3): 513-526, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27523270

RESUMEN

The kinases RIPK1 and RIPK3 and the pseudo-kinase MLKL have been identified as key regulators of the necroptotic cell death pathway, although a role for MLKL within the whole animal has not yet been established. Here, we have shown that MLKL deficiency rescued the embryonic lethality caused by loss of Caspase-8 or FADD. Casp8(-/-)Mlkl(-/-) and Fadd(-/-)Mlkl(-/-) mice were viable and fertile but rapidly developed severe lymphadenopathy, systemic autoimmune disease, and thrombocytopenia. These morbidities occurred more rapidly and with increased severity in Casp8(-/-)Mlkl(-/-) and Fadd(-/-)Mlkl(-/-) mice compared to Casp8(-/-)Ripk3(-/-) or Fadd(-/-)Ripk3(-/-) mice, respectively. These results demonstrate that MLKL is an essential effector of aberrant necroptosis in embryos caused by loss of Caspase-8 or FADD. Furthermore, they suggest that RIPK3 and/or MLKL may exert functions independently of necroptosis. It appears that non-necroptotic functions of RIPK3 contribute to the lymphadenopathy, autoimmunity, and excess cytokine production that occur when FADD or Caspase-8-mediated apoptosis is abrogated.


Asunto(s)
Apoptosis/fisiología , Enfermedades Autoinmunes/metabolismo , Muerte Celular/fisiología , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Caspasa 8/metabolismo , Ratones , Ratones Endogámicos C57BL , Necrosis/metabolismo
2.
Blood ; 139(17): 2673-2690, 2022 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-35245376

RESUMEN

The process of proplatelet formation (PPF) requires coordinated interaction between megakaryocytes (MKs) and the extracellular matrix (ECM), followed by a dynamic reorganization of the actin and microtubule cytoskeleton. Localized fluxes of intracellular calcium ions (Ca2+) facilitate MK-ECM interaction and PPF. Glutamate-gated N-methyl-D-aspartate receptor (NMDAR) is highly permeable to Ca2+. NMDAR antagonists inhibit MK maturation ex vivo; however, there are no in vivo data. Using the Cre-loxP system, we generated a platelet lineage-specific knockout mouse model of reduced NMDAR function in MKs and platelets (Pf4-Grin1-/- mice). Effects of NMDAR deletion were examined using well-established assays of platelet function and production in vivo and ex vivo. We found that Pf4-Grin1-/- mice had defects in megakaryopoiesis, thrombopoiesis, and platelet function, which manifested as reduced platelet counts, lower rates of platelet production in the immune model of thrombocytopenia, and prolonged tail bleeding time. Platelet activation was impaired to a range of agonists associated with reduced Ca2+ responses, including metabotropic like, and defective platelet spreading. MKs showed reduced colony and proplatelet formation. Impaired reorganization of intracellular F-actin and α-tubulin was identified as the main cause of reduced platelet function and production. Pf4-Grin1-/- MKs also had lower levels of transcripts encoding crucial ECM elements and enzymes, suggesting NMDAR signaling is involved in ECM remodeling. In summary, we provide the first genetic evidence that NMDAR plays an active role in platelet function and production. NMDAR regulates PPF through a mechanism that involves MK-ECM interaction and cytoskeletal reorganization. Our results suggest that NMDAR helps guide PPF in vivo.


Asunto(s)
Megacariocitos/metabolismo , Proteínas del Tejido Nervioso/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Trombocitopenia , Actinas/metabolismo , Animales , Plaquetas/metabolismo , Calcio , Ratones , Ratones Noqueados , Receptores de N-Metil-D-Aspartato/genética , Trombocitopenia/genética , Trombopoyesis/fisiología
3.
Blood ; 139(9): 1359-1373, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-34852174

RESUMEN

RNA processing is increasingly recognized as a critical control point in the regulation of different hematopoietic lineages including megakaryocytes responsible for the production of platelets. Platelets are anucleate cytoplasts that contain a rich repertoire of RNAs encoding proteins with essential platelet functions derived from the parent megakaryocyte. It is largely unknown how RNA binding proteins contribute to the development and functions of megakaryocytes and platelets. We show that serine-arginine-rich splicing factor 3 (SRSF3) is essential for megakaryocyte maturation and generation of functional platelets. Megakaryocyte-specific deletion of Srsf3 in mice led to macrothrombocytopenia characterized by megakaryocyte maturation arrest, dramatically reduced platelet counts, and abnormally large functionally compromised platelets. SRSF3 deficient megakaryocytes failed to reprogram their transcriptome during maturation and to load platelets with RNAs required for normal platelet function. SRSF3 depletion led to nuclear accumulation of megakaryocyte mRNAs, demonstrating that SRSF3 deploys similar RNA regulatory mechanisms in megakaryocytes as in other cell types. Our study further suggests that SRSF3 plays a role in sorting cytoplasmic megakaryocyte RNAs into platelets and demonstrates how SRSF3-mediated RNA processing forms a central part of megakaryocyte gene regulation. Understanding SRSF3 functions in megakaryocytes and platelets provides key insights into normal thrombopoiesis and platelet pathologies as SRSF3 RNA targets in megakaryocytes are associated with platelet diseases.


Asunto(s)
Plaquetas/metabolismo , Megacariocitos/metabolismo , ARN Mensajero , Factores de Empalme Serina-Arginina , Trombocitopenia , Trombopoyesis/genética , Animales , Ratones , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo , Trombocitopenia/genética , Trombocitopenia/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 41(3): 1167-1178, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33441028

RESUMEN

OBJECTIVE: People with diabetes are at a significantly higher risk of cardiovascular disease, in part, due to accelerated atherosclerosis. Diabetic subjects have increased number of platelets that are activated, more reactive, and respond suboptimally to antiplatelet therapies. We hypothesized that reducing platelet numbers by inducing their premature apoptotic death would decrease atherosclerosis. Approach and Results: This was achieved by targeting the antiapoptotic protein Bcl-xL (B-cell lymphoma-extra large; which is essential for platelet viability) via distinct genetic and pharmacological approaches. In the former, we transplanted bone marrow from mice carrying the Tyr15 to Cys loss of function allele of Bcl-x (known as Bcl-xPlt20) or wild-type littermate controls into atherosclerotic-prone Ldlr+/- mice made diabetic with streptozotocin and fed a Western diet. Reduced Bcl-xL function in hematopoietic cells significantly decreased platelet numbers, exclusive of other hematologic changes. This led to a significant reduction in atherosclerotic lesion formation in Bcl-xPlt20 bone marrow transplanted Ldlr+/- mice. To assess the potential therapeutic relevance of reducing platelets in atherosclerosis, we next targeted Bcl-xL with a pharmacological strategy. This was achieved by low-dose administration of the BH3 (B-cell lymphoma-2 homology domain 3) mimetic, ABT-737 triweekly, in diabetic Apoe-/- mice for the final 6 weeks of a 12-week study. ABT-737 normalized platelet numbers along with platelet and leukocyte activation to that of nondiabetic controls, significantly reducing atherosclerosis while promoting a more stable plaque phenotype. CONCLUSIONS: These studies suggest that selectively reducing circulating platelets, by targeting Bcl-xL to promote platelet apoptosis, can reduce atherosclerosis and lower cardiovascular disease risk in diabetes. Graphic Abstract: A graphic abstract is available for this article.


Asunto(s)
Aterosclerosis/sangre , Aterosclerosis/complicaciones , Plaquetas/patología , Angiopatías Diabéticas/sangre , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Aterosclerosis/prevención & control , Compuestos de Bifenilo/administración & dosificación , Plaquetas/efectos de los fármacos , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/complicaciones , Femenino , Humanos , Leucocitos/patología , Leucocitos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nitrofenoles/administración & dosificación , Piperazinas/administración & dosificación , Recuento de Plaquetas , Receptores de LDL/deficiencia , Receptores de LDL/genética , Factores de Riesgo , Sulfonamidas/administración & dosificación
5.
Immunity ; 36(4): 646-57, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22483802

RESUMEN

The immune system must distinguish viable cells from cells damaged by physical and infective processes. The damaged cell-recognition molecule Clec9A is expressed on the surface of the mouse and human dendritic cell subsets specialized for the uptake and processing of material from dead cells. Clec9A recognizes a conserved component within nucleated and nonnucleated cells, exposed when cell membranes are damaged. We have identified this Clec9A ligand as a filamentous form of actin in association with particular actin-binding domains of cytoskeletal proteins. We have determined the crystal structure of the human CLEC9A C-type lectin domain and propose a functional dimeric structure with conserved tryptophans in the ligand recognition site. Mutation of these residues ablated CLEC9A binding to damaged cells and to the isolated ligand complexes. We propose that Clec9A provides targeted recruitment of the adaptive immune system during infection and can also be utilized to enhance immune responses generated by vaccines.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Mitogénicos/metabolismo , Actinas/metabolismo , Inmunidad Adaptativa , Animales , Sitios de Unión , Línea Celular , Membrana Celular/metabolismo , Células Dendríticas/citología , Femenino , Humanos , Lectinas Tipo C/química , Lectinas Tipo C/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Estructura Secundaria de Proteína , Receptores Inmunológicos/genética , Receptores Mitogénicos/química , Receptores Mitogénicos/genética , Espectrina/metabolismo
6.
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
7.
Int J Mol Sci ; 21(20)2020 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-33066573

RESUMEN

Blood platelets have important roles in haemostasis, where they quickly stop bleeding in response to vascular damage. They have also recognised functions in thrombosis, immunity, antimicrobal defense, cancer growth and metastasis, tumour angiogenesis, lymphangiogenesis, inflammatory diseases, wound healing, liver regeneration and neurodegeneration. Their brief life span in circulation is strictly controlled by intrinsic apoptosis, where the prosurvival Bcl-2 family protein, Bcl-xL, has a major role. Blood platelets are produced by large polyploid precursor cells, megakaryocytes, residing mainly in the bone marrow. Together with Mcl-1, Bcl-xL regulates megakaryocyte survival. This review describes megakaryocyte maturation and survival, platelet production, platelet life span and diseases of abnormal platelet number with a focus on the role of Bcl-xL during these processes.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/metabolismo , Plaquetas/citología , Proteína bcl-X/metabolismo , Animales , Apoptosis , Trastornos de las Plaquetas Sanguíneas/genética , Plaquetas/metabolismo , Humanos , Megacariocitos/citología , Megacariocitos/metabolismo , Trombopoyesis , Proteína bcl-X/genética
8.
Blood ; 126(6): 807-16, 2015 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-25995324

RESUMEN

The thrombopoietic environment of the neonate is established during prenatal life; therefore, a comprehensive understanding of platelet-forming cell development during embryogenesis is critical to understanding the etiology of early-onset thrombocytopenia. The recent discovery that the first platelet-forming cells of the conceptus are not megakaryocytes (MKs) but diploid platelet-forming cells (DPFCs) revealed a previously unappreciated complexity in thrombopoiesis. This raises important questions, including the following. When do conventional MKs appear? Do pathogenic genetic lesions of adult MKs affect DPFCs? What role does myeloproliferative leukemia virus (MPL), a key regulator of adult megakaryopoiesis, play in prenatal platelet-forming lineages? We performed a comprehensive study to determine the spatial and temporal appearance of prenatal platelet-forming lineages. We demonstrate that DPFCs originate in the yolk sac and then rapidly migrate to other extra- and intraembryonic tissues. Using gene disruption models of Gata1 and Nfe2, we demonstrate that perturbing essential adult MK genes causes an analogous phenotype in the early embryo before the onset of hematopoietic stem/progenitor cell-driven (definitive) hematopoiesis. Finally, we present the surprising finding that DPFC and MK commitment from their respective precursors is MPL independent in vivo but that completion of MK differentiation and establishment of the prenatal platelet mass is dependent on MPL expression.


Asunto(s)
Plaquetas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Megacariocitos/metabolismo , Receptores de Trombopoyetina/genética , Trombopoyesis/genética , Saco Vitelino/metabolismo , Animales , Plaquetas/citología , Diferenciación Celular , Linaje de la Célula/genética , Desarrollo Embrionario/genética , Factor de Transcripción GATA1/deficiencia , Factor de Transcripción GATA1/genética , Eliminación de Gen , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Megacariocitos/citología , Ratones , Ratones Noqueados , Subunidad p45 del Factor de Transcripción NF-E2/deficiencia , Subunidad p45 del Factor de Transcripción NF-E2/genética , Receptores de Trombopoyetina/metabolismo , Transcripción Genética , Saco Vitelino/citología , Saco Vitelino/crecimiento & desarrollo
9.
Blood ; 126(2): 167-75, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26036803

RESUMEN

Polycomb repressive complex 2 (PRC2) is a chromatin modifier that regulates stem cells in embryonic and adult tissues. Loss-of-function studies of PRC2 components have been complicated by early embryonic dependence on PRC2 activity and the partial functional redundancy of enhancer of zeste homolog 1 (Ezh1) and enhancer of zeste homolog 2 (Ezh2), which encode the enzymatic component of PRC2. Here, we investigated the role of PRC2 in hematopoiesis by conditional deletion of suppressor of zeste 12 protein homolog (Suz12), a core component of PRC2. Complete loss of Suz12 resulted in failure of hematopoiesis, both in the embryo and the adult, with a loss of maintenance of hematopoietic stem cells (HSCs). In contrast, partial loss of PRC2 enhanced HSC self-renewal. Although Suz12 was required for lymphoid development, deletion in individual blood cell lineages revealed that it was dispensable for the development of granulocytic, monocytic, and megakaryocytic cells. Collectively, these data reveal the multifaceted role of PRC2 in hematopoiesis, with divergent dose-dependent effects in HSC and distinct roles in maturing blood cells. Because PRC2 is a potential target for cancer therapy, the significant consequences of modest changes in PRC2 activity, as well as the cell and developmental stage-specific effects, will need to be carefully considered in any therapeutic context.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Linfopoyesis/genética , Complejo Represivo Polycomb 2/fisiología , Animales , Proliferación Celular/genética , Células Cultivadas , Feto/inmunología , Feto/fisiología , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Complejo Represivo Polycomb 2/genética
10.
Platelets ; 28(4): 342-353, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27494300

RESUMEN

Extracellular proteolysis of platelet plasma membrane proteins is an event that ensues platelet activation. Shedding of surface receptors such as glycoprotein (GP) Ibα, GPV and GPVI as well as externalized proteins P-selectin and CD40L releases soluble ectodomain fragments that are subsequently detectable in plasma. This results in the irreversible functional downregulation of platelet receptor-mediated adhesive interactions and the generation of biologically active fragments. In this review, we describe molecular insights into the regulation of platelet receptor and ligand shedding in health and disease. The scope of this review is specially focused on GPIbα, GPV, GPVI, P-selectin and CD40L where we: (1) describe the basic physiological regulation of expression and shedding of these proteins in hemostasis illustrate alterations in receptor expression during (2) apoptosis and (3) ex vivo storage relevant for blood banking purposes; (4) discuss considerations to be made when analyzing and interpreting shedding of platelet membrane proteins and finally; (5) collate clinical evidence that quantify these platelet proteins during disease.


Asunto(s)
Plaquetas/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Enfermedad , Humanos , Selectina-P/sangre , Activación Plaquetaria , Agregación Plaquetaria
11.
Proc Natl Acad Sci U S A ; 111(16): 5884-9, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24711413

RESUMEN

Thrombopoietin (TPO) acting via its receptor, the cellular homologue of the myeloproliferative leukemia virus oncogene (Mpl), is the major cytokine regulator of platelet number. To precisely define the role of specific hematopoietic cells in TPO-dependent hematopoiesis, we generated mice that express the Mpl receptor normally on stem/progenitor cells but lack expression on megakaryocytes and platelets (Mpl(PF4cre/PF4cre)). Mpl(PF4cre/PF4cre) mice displayed profound megakaryocytosis and thrombocytosis with a remarkable expansion of megakaryocyte-committed and multipotential progenitor cells, the latter displaying biological responses and a gene expression signature indicative of chronic TPO overstimulation as the underlying causative mechanism, despite a normal circulating TPO level. Thus, TPO signaling in megakaryocytes is dispensable for platelet production; its key role in control of platelet number is via generation and stimulation of the bipotential megakaryocyte precursors. Nevertheless, Mpl expression on megakaryocytes and platelets is essential to prevent megakaryocytosis and myeloproliferation by restricting the amount of TPO available to stimulate the production of megakaryocytes from the progenitor cell pool.


Asunto(s)
Plaquetas/metabolismo , Megacariocitos/metabolismo , Células Mieloides/citología , Receptores de Trombopoyetina/metabolismo , Trombopoyesis , Animales , Antígenos CD34/metabolismo , Plaquetas/citología , Compartimento Celular , Proliferación Celular , Células Clonales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Marcación de Gen , Sitios Genéticos/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Integrasas/metabolismo , Megacariocitos/citología , Ratones , Modelos Biológicos , Células Mieloides/metabolismo , Trombocitosis , Trombopoyetina/genética , Trombopoyetina/metabolismo , Transcripción Genética
12.
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
13.
Blood ; 124(17): 2725-9, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25079356

RESUMEN

In this study, we test the assumption that the hematopoietic progenitor/colony-forming cells of the embryonic yolk sac (YS), which are endowed with megakaryocytic potential, differentiate into the first platelet-forming cells in vivo. We demonstrate that from embryonic day (E) 8.5 all megakaryocyte (MK) colony-forming cells belong to the conventional hematopoietic progenitor cell (HPC) compartment. Although these cells are indeed capable of generating polyploid MKs, they are not the source of the first platelet-forming cells. We show that proplatelet formation first occurs in a unique and previously unrecognized lineage of diploid platelet-forming cells, which develop within the YS in parallel to HPCs but can be specified in the E8.5 Runx1-null embryo despite the absence of the progenitor cell lineage.


Asunto(s)
Linaje de la Célula/genética , Diploidia , Embrión de Mamíferos/metabolismo , Células Progenitoras de Megacariocitos/metabolismo , Megacariocitos/metabolismo , Poliploidía , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Regulación del Desarrollo de la Expresión Génica , Células Progenitoras de Megacariocitos/citología , Megacariocitos/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Tiempo , Transcriptoma , Saco Vitelino/citología , Saco Vitelino/embriología , Saco Vitelino/metabolismo
14.
Platelets ; 27(6): 497-504, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27100842

RESUMEN

The lifespan of platelets in circulation is brief, close to 10 days in humans and 5 days in mice. Bone marrow residing megakaryocytes produce around 100 billion platelets per day. In a healthy individual, the majority of platelets are not consumed by hemostatic processes, but rather their lifespan is controlled by programmed cell death, a canonical intrinsic apoptosis program. In the last decade, insights from genetically manipulated mouse models and pharmacological developments have helped to define the components of the intrinsic, or mitochondrial, apoptosis pathway that controls platelet lifespan. This review focuses on the molecular regulation of apoptosis in platelet survival, reviews thrombocytopenic conditions linked to enhanced platelet death, examines implications of chemotherapy-induced thrombocytopenia through apoptosis-inducing drugs in cancer therapy as well as discusses ex vivo aging of platelets.


Asunto(s)
Apoptosis , Plaquetas/fisiología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Plaquetas/efectos de los fármacos , Conservación de la Sangre , Caspasas/metabolismo , Regulación de la Expresión Génica , Hemostasis , Humanos , Megacariocitos/citología , Megacariocitos/efectos de los fármacos , Megacariocitos/fisiología , Fosfatidilserinas/metabolismo , Fosfatidilserinas/farmacología , Transfusión de Plaquetas , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/etiología , Trombocitopenia/metabolismo
15.
Proc Natl Acad Sci U S A ; 110(7): 2599-604, 2013 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-23349374

RESUMEN

Dysregulation of the "intrinsic" apoptotic pathway is associated with the development of cancer and autoimmune disease. Bak and Bax are two proapoptotic members of the Bcl-2 protein family with overlapping, essential roles in the intrinsic apoptotic pathway. Their activity is critical for the control of cell survival during lymphocyte development and homeostasis, best demonstrated by defects in thymic T-cell differentiation and peripheral lymphoid homeostasis caused by their combined loss. Because most bak(-/-)bax(-/-) mice die perinatally, the roles of Bax and Bak in immunological tolerance and prevention of autoimmune disease remain unclear. We show that mice reconstituted with a Bak/Bax doubly deficient hematopoietic compartment develop a fatal systemic lupus erythematosus-like autoimmune disease characterized by hypergammaglobulinemia, autoantibodies, lymphadenopathy, glomerulonephritis, and vasculitis. Importantly, these mice also develop a multiorgan autoimmune disease with autoantibodies against most solid glandular structures and evidence of glandular atrophy and necrotizing vasculitis. Interestingly, similar albeit less severe pathology was observed in mice containing a hematopoietic compartment deficient for only Bak, a phenotype reminiscent of the disease seen in patients with point mutations in BAK. These studies demonstrate a critical role for Bak and an ancillary role for Bax in safeguarding immunological tolerance and prevention of autoimmune disease. This suggests that direct activators of the intrinsic apoptotic pathway, such as BH3 mimetics, may be useful for treatment of diverse autoimmune diseases.


Asunto(s)
Apoptosis/inmunología , Enfermedades Autoinmunes/inmunología , Proteína Destructora del Antagonista Homólogo bcl-2/inmunología , Proteína X Asociada a bcl-2/inmunología , Animales , Autoanticuerpos/inmunología , Enfermedades Autoinmunes/patología , Western Blotting , Quimiocinas/sangre , Cruzamientos Genéticos , Citocinas/sangre , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnicas Histológicas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Proteína Destructora del Antagonista Homólogo bcl-2/deficiencia , Proteína X Asociada a bcl-2/deficiencia
16.
Blood ; 119(18): 4283-90, 2012 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-22294729

RESUMEN

Apoptotic caspases, including caspase-9, are thought to facilitate platelet shedding by megakaryocytes. They are known to be activated during platelet apoptosis, and have also been implicated in platelet hemostatic responses. However, the precise requirement for, and the regulation of, apoptotic caspases have never been defined in either megakaryocytes or platelets. To establish the role of caspases in platelet production and function, we generated mice lacking caspase-9 in their hematopoietic system. We demonstrate that both megakaryocytes and platelets possess a functional apoptotic caspase cascade downstream of Bcl-2 family-mediated mitochondrial damage. Caspase-9 is the initiator caspase, and its loss blocks effector caspase activation. Surprisingly, steady-state thrombopoiesis is unperturbed in the absence of caspase-9, indicating that the apoptotic caspase cascade is not required for platelet production. In platelets, loss of caspase-9 confers resistance to the BH3 mimetic ABT-737, blocking phosphatidylserine (PS) exposure and delaying ABT-737-induced thrombocytopenia in vivo. Despite this, steady-state platelet lifespan is normal. Casp9(-/-) platelets are fully capable of physiologic hemostatic responses and functional regulation of adhesive integrins in response to agonist. These studies demonstrate that the apoptotic caspase cascade is required for the efficient death of megakaryocytes and platelets, but is dispensable for their generation and function.


Asunto(s)
Apoptosis/fisiología , Plaquetas/citología , Caspasa 9/fisiología , Megacariocitos/citología , Trombopoyesis/fisiología , Animales , Compuestos de Bifenilo/farmacología , Compuestos de Bifenilo/toxicidad , Plaquetas/enzimología , Caspasa 9/deficiencia , Caspasa 9/genética , Linaje de la Célula , Hemostasis/efectos de los fármacos , Hemostasis/fisiología , Hirudinas/farmacología , Hígado/embriología , Trasplante de Hígado , Megacariocitos/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nitrofenoles/farmacología , Nitrofenoles/toxicidad , Piperazinas/farmacología , Piperazinas/toxicidad , Activación Plaquetaria/efectos de los fármacos , Activación Plaquetaria/fisiología , Quimera por Radiación , Sulfonamidas/farmacología , Sulfonamidas/toxicidad , Trombocitopenia/inducido químicamente , Proteína X Asociada a bcl-2/deficiencia
17.
Blood ; 119(24): 5850-8, 2012 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-22374700

RESUMEN

Mature megakaryocytes depend on the function of Bcl-x(L), a member of the Bcl-2 family of prosurvival proteins, to proceed safely through the process of platelet shedding. Despite this, loss of Bcl-x(L) does not prevent the growth and maturation of megakaryocytes, suggesting redundancy with other prosurvival proteins. We therefore generated mice with a megakaryocyte-specific deletion of Mcl-1, which is known to be expressed in megakaryocytes. Megakaryopoiesis, platelet production, and platelet lifespan were unperturbed in Mcl-1(Pf4Δ/Pf4Δ) animals. However, treatment with ABT-737, a BH3 mimetic compound that inhibits the prosurvival proteins Bcl-2, Bcl-x(L), and Bcl-w resulted in the complete ablation of megakaryocytes and platelets. Genetic deletion of both Mcl-1 and Bcl-x(L) in megakaryocytes resulted in preweaning lethality. Megakaryopoiesis in Bcl-x(Pf4Δ/Pf4Δ) Mcl-1(Pf4Δ/Pf4Δ) embryos was severely compromised, and these animals exhibited ectopic bleeding. Our studies indicate that the combination of Bcl-x(L) and Mcl-1 is essential for the viability of the megakaryocyte lineage.


Asunto(s)
Megacariocitos/metabolismo , Megacariocitos/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína bcl-X/metabolismo , Alelos , Animales , Compuestos de Bifenilo/administración & dosificación , Compuestos de Bifenilo/farmacología , Recuento de Células Sanguíneas , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Plaquetas/patología , Recuento de Células , Muerte Celular/efectos de los fármacos , Tamaño de la Célula , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/patología , Feto/efectos de los fármacos , Feto/metabolismo , Feto/patología , Eliminación de Gen , Hemorragia/patología , Hígado/efectos de los fármacos , Hígado/embriología , Hígado/metabolismo , Hígado/patología , Vasos Linfáticos/efectos de los fármacos , Vasos Linfáticos/patología , Megacariocitos/efectos de los fármacos , Megacariocitos/ultraestructura , Ratones , Ratones Endogámicos C57BL , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Nitrofenoles/administración & dosificación , Nitrofenoles/farmacología , Especificidad de Órganos/efectos de los fármacos , Piperazinas/administración & dosificación , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/deficiencia , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Trombopoyesis/efectos de los fármacos
18.
Blood ; 119(5): 1263-73, 2012 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-22101895

RESUMEN

When refrigerated platelets are rewarmed, they secrete active sialidases, including the lysosomal sialidase Neu1, and express surface Neu3 that remove sialic acid from platelet von Willebrand factor receptor (VWFR), specifically the GPIbα subunit. The recovery and circulation of refrigerated platelets is greatly improved by storage in the presence of inhibitors of sialidases. Desialylated VWFR is also a target for metalloproteinases (MPs), because GPIbα and GPV are cleaved from the surface of refrigerated platelets. Receptor shedding is inhibited by the MP inhibitor GM6001 and does not occur in Adam17(ΔZn/ΔZn) platelets expressing inactive ADAM17. Critically, desialylation in the absence of MP-mediated receptor shedding is sufficient to cause the rapid clearance of platelets from circulation. Desialylation of platelet VWFR therefore triggers platelet clearance and primes GPIbα and GPV for MP-dependent cleavage.


Asunto(s)
Plaquetas/fisiología , Glicoproteínas de Membrana/metabolismo , Metaloproteasas/metabolismo , Neuraminidasa/metabolismo , Refrigeración , Proteínas ADAM/metabolismo , Proteínas ADAM/fisiología , Proteína ADAM17 , Animales , Plaquetas/metabolismo , Conservación de la Sangre/métodos , Activación Enzimática , Glicosilación , Humanos , Masculino , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/fisiología , Metaloproteasas/fisiología , Ratones , Ratones Endogámicos C57BL , Neuraminidasa/fisiología , Complejo GPIb-IX de Glicoproteína Plaquetaria , Procesamiento Proteico-Postraduccional/fisiología , Proteolisis , Refrigeración/métodos , Factor de von Willebrand/metabolismo
19.
Blood ; 120(3): 626-35, 2012 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-22613794

RESUMEN

Platelets are megakaryocyte subfragments that participate in hemostatic and host defense reactions and deliver pro- and antiangiogenic factors throughout the vascular system. Although they are anucleated cells that lack a complex secretory apparatus with distinct Golgi/endoplasmic reticulum compartments, past studies have shown that platelets have glycosyltransferase activities. In the present study, we show that members of 3 distinct glycosyltransferase families are found within and on the surface of platelets. Immunocytology and flow cytometry results indicated that megakaryocytes package these Golgi-derived glycosyltransferases into vesicles that are sent via proplatelets to nascent platelets, where they accumulate. These glycosyltransferases are active, and intact platelets glycosylate large exogenous substrates. Furthermore, we show that activation of platelets results in the release of soluble glycosyltransferase activities and that platelets contain sufficient levels of sugar nucleotides for detection of glycosylation of exogenously added substrates. Therefore, the results of the present study show that blood platelets are a rich source of both glycosyltransferases and donor sugar substrates that can be released to function in the extracellular space. This platelet-glycosylation machinery offers a pathway to a simple glycoengineering strategy improving storage of platelets and may serve hitherto unknown biologic functions.


Asunto(s)
Plaquetas/enzimología , Glicosiltransferasas/metabolismo , Megacariocitos/enzimología , Familia de Multigenes/fisiología , Animales , Galactosiltransferasas/genética , Galactosiltransferasas/metabolismo , Glicosilación , Aparato de Golgi/enzimología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , N-Acetilgalactosaminiltransferasas/genética , N-Acetilgalactosaminiltransferasas/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Plasma Rico en Plaquetas/citología , Sialiltransferasas/genética , Sialiltransferasas/metabolismo , Especificidad por Sustrato/fisiología , Polipéptido N-Acetilgalactosaminiltransferasa
20.
Am J Hematol ; 89(12): E228-34, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25132654

RESUMEN

Platelet survival depends upon mediators of apoptosis e.g., Bcl-xL, Bax, and Bak, which are regulated by thrombopoietin (TPO)-mediated AKT signaling. Thrombopoietin receptor (TPO-R) signaling might decrease platelet and/or megakaryocyte apoptosis and increase the platelet count. This study therefore explored anti-apoptotic effects of TPO-R-agonists in vivo on platelets of patients with immune thrombocytopenia. Patients received eltrombopag or romiplostim for two weeks. Total, immature, and large platelet counts were assessed as were Bcl-xL inhibitor assay; Bcl-xL Western blot; and flow cytometric (FACS) analysis of the AKT-signaling pathway. Eight/ten patients had platelet responses to eltrombopag and all three to romiplostim. Platelet sensitivity to apoptosis by Bcl-xL inhibition was greater in pretreatment patients than controls. This sensitivity normalized after one week of therapy, but surprisingly returned to pretreatment levels at week two. FACS analysis revealed increased AKT-pathway signaling after one week, followed by a decrease at week two. Platelet counts correlated with the Bcl-xL /Bak ratio. Platelet survival may be enhanced by TPO-R-agonists as a transient decrease in platelet sensitivity to apoptosis was accompanied by transient activation of AKT. However, this mechanism has only a short-lived effect. Megakaryocytes and platelets already present at the start of TPO-R-agonist treatment appear to respond differently than those generated de novo.


Asunto(s)
Benzoatos/uso terapéutico , Plaquetas/efectos de los fármacos , Hidrazinas/uso terapéutico , Megacariocitos/efectos de los fármacos , Púrpura Trombocitopénica Idiopática/tratamiento farmacológico , Pirazoles/uso terapéutico , Receptores Fc/uso terapéutico , Receptores de Trombopoyetina/agonistas , Proteínas Recombinantes de Fusión/uso terapéutico , Trombopoyetina/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Plaquetas/metabolismo , Plaquetas/patología , Estudios de Casos y Controles , Enfermedad Crónica , Regulación de la Expresión Génica , Humanos , Megacariocitos/metabolismo , Megacariocitos/patología , Púrpura Trombocitopénica Idiopática/genética , Púrpura Trombocitopénica Idiopática/metabolismo , Púrpura Trombocitopénica Idiopática/patología , Receptores de Trombopoyetina/genética , Receptores de Trombopoyetina/metabolismo , Transducción de Señal , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA