Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 145
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 138(14): 1211-1224, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34115843

RESUMEN

Megakaryocytes (MKs), the platelet progenitor cells, play important roles in hematopoietic stem cell (HSC) maintenance and immunity. However, it is not known whether these diverse programs are executed by a single population or by distinct subsets of cells. Here, we manually isolated primary CD41+ MKs from the bone marrow (BM) of mice and human donors based on ploidy (2N-32N) and performed single-cell RNA sequencing analysis. We found that cellular heterogeneity existed within 3 distinct subpopulations that possess gene signatures related to platelet generation, HSC niche interaction, and inflammatory responses. In situ immunostaining of mouse BM demonstrated that platelet generation and the HSC niche-related MKs were in close physical proximity to blood vessels and HSCs, respectively. Proplatelets, which could give rise to platelets under blood shear forces, were predominantly formed on a platelet generation subset. Remarkably, the inflammatory responses subpopulation, consisting generally of low-ploidy LSP1+ and CD53+ MKs (≤8N), represented ∼5% of total MKs in the BM. These MKs could specifically respond to pathogenic infections in mice. Rapid expansion of this population was accompanied by strong upregulation of a preexisting PU.1- and IRF-8-associated monocytic-like transcriptional program involved in pathogen recognition and clearance as well as antigen presentation. Consistently, isolated primary CD53+ cells were capable of engulfing and digesting bacteria and stimulating T cells in vitro. Together, our findings uncover new molecular, spatial, and functional heterogeneity within MKs in vivo and demonstrate the existence of a specialized MK subpopulation that may act as a new type of immune cell.


Asunto(s)
Ratones/genética , Análisis de la Célula Individual , Trombopoyesis , Transcriptoma , Animales , Células Cultivadas , Femenino , Humanos , Masculino , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones/fisiología , Ratones Endogámicos C57BL , Glicoproteína IIb de Membrana Plaquetaria/análisis , Glicoproteína IIb de Membrana Plaquetaria/genética , Ploidias
2.
Blood ; 137(16): 2139-2151, 2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33667305

RESUMEN

We studied a subset of hematopoietic stem cells (HSCs) that are defined by elevated expression of CD41 (CD41hi) and showed bias for differentiation toward megakaryocytes (Mks). Mouse models of myeloproliferative neoplasms (MPNs) expressing JAK2-V617F (VF) displayed increased frequencies and percentages of the CD41hi vs CD41lo HSCs compared with wild-type controls. An increase in CD41hi HSCs that correlated with JAK2-V617F mutant allele burden was also found in bone marrow from patients with MPN. CD41hi HSCs produced a higher number of Mk-colonies of HSCs in single-cell cultures in vitro, but showed reduced long-term reconstitution potential compared with CD41lo HSCs in competitive transplantations in vivo. RNA expression profiling showed an upregulated cell cycle, Myc, and oxidative phosphorylation gene signatures in CD41hi HSCs, whereas CD41lo HSCs showed higher gene expression of interferon and the JAK/STAT and TNFα/NFκB signaling pathways. Higher cell cycle activity and elevated levels of reactive oxygen species were confirmed in CD41hi HSCs by flow cytometry. Expression of Epcr, a marker for quiescent HSCs inversely correlated with expression of CD41 in mice, but did not show such reciprocal expression pattern in patients with MPN. Treatment with interferon-α further increased the frequency and percentage of CD41hi HSCs and reduced the number of JAK2-V617F+ HSCs in mice and patients with MPN. The shift toward the CD41hi subset of HSCs by interferon-α provides a possible mechanism of how interferon-α preferentially targets the JAK2 mutant clone.


Asunto(s)
Interferón-alfa/uso terapéutico , Janus Quinasa 2/genética , Megacariocitos/metabolismo , Trastornos Mieloproliferativos/genética , Animales , Técnicas de Sustitución del Gen , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Megacariocitos/citología , Ratones , Ratones Transgénicos , Trastornos Mieloproliferativos/tratamiento farmacológico , Glicoproteína IIb de Membrana Plaquetaria/genética , Mutación Puntual/efectos de los fármacos
3.
Blood ; 138(15): 1359-1372, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34375384

RESUMEN

The αIIbß3 integrin receptor coordinates platelet adhesion, activation, and mechanosensing in thrombosis and hemostasis. Using differential cysteine alkylation and mass spectrometry, we have identified a disulfide bond in the αIIb subunit linking cysteines 490 and 545 that is missing in ∼1 in 3 integrin molecules on the resting and activated human platelet surface. This alternate covalent form of αIIbß3 is predetermined as it is also produced by human megakaryoblasts and baby hamster kidney fibroblasts transfected with recombinant integrin. From coimmunoprecipitation experiments, the alternate form selectively partitions into focal adhesions on the activated platelet surface. Its function was evaluated in baby hamster kidney fibroblast cells expressing a mutant integrin with an ablated C490-C545 disulfide bond. The disulfide mutant integrin has functional outside-in signaling but extended residency time in focal adhesions due to a reduced rate of clathrin-mediated integrin internalization and recycling, which is associated with enhanced affinity of the αIIb subunit for clathrin adaptor protein 2. Molecular dynamics simulations indicate that the alternate covalent form of αIIb requires higher forces to transition from bent to open conformational states that is in accordance with reduced affinity for fibrinogen and activation by manganese ions. These findings indicate that the αIIbß3 integrin receptor is produced in various covalent forms that have different cell surface distribution and function. The C490, C545 cysteine pair is conserved across all 18 integrin α subunits, and the disulfide bond in the αV and α2 subunits in cultured cells is similarly missing, suggesting that the alternate integrin form and function are also conserved.


Asunto(s)
Adhesiones Focales/metabolismo , Integrina beta3/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Animales , Línea Celular , Cricetinae , Disulfuros/análisis , Adhesiones Focales/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina beta3/química , Integrina beta3/genética , Simulación de Dinámica Molecular , Mutación , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/química , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/genética , Glicoproteína IIb de Membrana Plaquetaria/química , Glicoproteína IIb de Membrana Plaquetaria/genética
4.
Int J Mol Sci ; 23(4)2022 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-35216179

RESUMEN

HLJ1 (also called DNAJB4) is a member of the DNAJ/Hsp40 family and plays an important role in regulating protein folding and activity. However, there is little information about the role of HLJ1 in the regulation of physiological function. In this study, we investigated the role of HLJ1 in blood coagulation using wild-type C57BL/6 mice and HLJ1-null (HLJ1-/-) mice. Western blot analysis and immunohistochemistry were used to assess the expression and distribution of HLJ1 protein, respectively. The tail bleeding assay was applied to assess the bleeding time and blood loss. A coagulation test was used for measuring the activity of extrinsic, intrinsic and common coagulation pathways. Thromboelastography was used to measure the coagulation parameters in the progression of blood clot formation. The results showed that HLJ1 was detectable in plasma and bone marrow. The distribution of HLJ1 was co-localized with CD41, the marker of platelets and megakaryocytes. However, genetic deletion of HLJ1 did not alter blood loss and the activity of extrinsic and intrinsic coagulation pathways, as well as blood clot formation, compared to wild-type mice. Collectively, these findings suggest that, although HLJ1 appears in megakaryocytes and platelets, it may not play a role in the function of blood coagulation under normal physiological conditions.


Asunto(s)
Coagulación Sanguínea/genética , Proteínas del Choque Térmico HSP40/genética , Proteínas del Choque Térmico HSP40/metabolismo , Animales , Biomarcadores/metabolismo , Plaquetas/metabolismo , Eliminación de Gen , Masculino , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Glicoproteína IIb de Membrana Plaquetaria/genética
5.
Int J Mol Sci ; 23(2)2022 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-35055046

RESUMEN

Integrin αIIbß3, a glycoprotein complex expressed at the platelet surface, is involved in platelet aggregation and contributes to primary haemostasis. Several integrin αIIbß3 polymorphisms prevent the aggregation that causes haemorrhagic syndromes, such as Glanzmann thrombasthenia (GT). Access to 3D structure allows understanding the structural effects of polymorphisms related to GT. In a previous analysis using Molecular Dynamics (MD) simulations of αIIbCalf-1 domain structure, it was observed that GT associated with single amino acid variation affects distant loops, but not the mutated position. In this study, experiments are extended to Calf-1, Thigh, and Calf-2 domains. Two loops in Calf-2 are unstructured and therefore are modelled expertly using biophysical restraints. Surprisingly, MD revealed the presence of rigid zones in these loops. Detailed analysis with structural alphabet, the Proteins Blocks (PBs), allowed observing local changes in highly flexible regions. The variant P741R located at C-terminal of Calf-1 revealed that the Calf-2 presence did not affect the results obtained with isolated Calf-1 domain. Simulations for Calf-1 + Calf-2, and Thigh + Calf-1 variant systems are designed to comprehend the impact of five single amino acid variations in these domains. Distant conformational changes are observed, thus highlighting the potential role of allostery in the structural basis of GT.


Asunto(s)
Mutación Missense , Glicoproteína IIb de Membrana Plaquetaria/química , Glicoproteína IIb de Membrana Plaquetaria/genética , Dominios y Motivos de Interacción de Proteínas/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Modelos Moleculares , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Conformación Proteica , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Relación Estructura-Actividad
6.
Int J Mol Sci ; 21(3)2020 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-32024018

RESUMEN

Megakaryopoiesis is the process during which megakaryoblasts differentiate to polyploid megakaryocytes that can subsequently shed thousands of platelets in the circulation. Megakaryocytes accumulate mRNA during their maturation, which is required for the correct spatio-temporal production of cytoskeletal proteins, membranes and platelet-specific granules, and for the subsequent shedding of thousands of platelets per cell. Gene expression profiling identified the RNA binding protein ATAXIN2 (ATXN2) as a putative novel regulator of megakaryopoiesis. ATXN2 expression is high in CD34+/CD41+ megakaryoblasts and sharply decreases upon maturation to megakaryocytes. ATXN2 associates with DDX6 suggesting that it may mediate repression of mRNA translation during early megakaryopoiesis. Comparative transcriptome and proteome analysis on megakaryoid cells (MEG-01) with differential ATXN2 expression identified ATXN2 dependent gene expression of mRNA and protein involved in processes linked to hemostasis. Mice deficient for Atxn2 did not display differences in bleeding times, but the expression of key surface receptors on platelets, such as ITGB3 (carries the CD61 antigen) and CD31 (PECAM1), was deregulated and platelet aggregation upon specific triggers was reduced.


Asunto(s)
Ataxina-2/genética , Perfilación de la Expresión Génica/métodos , Células Progenitoras de Megacariocitos/citología , Animales , Antígenos CD34/genética , Ataxina-2/metabolismo , Diferenciación Celular , Línea Celular , ARN Helicasas DEAD-box/genética , Regulación de la Expresión Génica , Humanos , Ratones , Glicoproteína IIb de Membrana Plaquetaria/genética , Proteínas Proto-Oncogénicas/genética
7.
Int J Mol Sci ; 21(1)2020 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-31948107

RESUMEN

Remodeling of the actin cytoskeleton is one of the critical events that allows platelets to undergo morphological and functional changes in response to receptor-mediated signaling cascades. Coronins are a family of evolutionarily conserved proteins implicated in the regulation of the actin cytoskeleton, represented by the abundant coronins 1, 2, and 3 and the less abundant coronin 7 in platelets, but their functions in these cells are poorly understood. A recent report revealed impaired agonist-induced actin polymerization and cofilin phosphoregulation and altered thrombus formation in vivo as salient phenotypes in the absence of an overt hemostasis defect in vivo in a knockout mouse model of coronin 1. Here we show that the absence of coronin 1 is associated with impaired translocation of integrin ß2 to the platelet surface upon stimulation with thrombin while morphological and functional alterations, including defects in Arp2/3 complex localization and cAMP-dependent signaling, are absent. Our results suggest a large extent of functional overlap among coronins 1, 2, and 3 in platelets, while aspects like integrin ß2 translocation are specifically or predominantly dependent on coronin 1.


Asunto(s)
Plaquetas/metabolismo , Cadenas beta de Integrinas/metabolismo , Proteínas de Microfilamentos/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Animales , Colágeno/farmacología , AMP Cíclico/metabolismo , Epoprostenol/farmacología , Integrina alfa2/genética , Integrina alfa2/metabolismo , Cadenas beta de Integrinas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Glicoproteína IIb de Membrana Plaquetaria/genética , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/genética , Unión Proteica , Transporte de Proteínas , Trombina/farmacología
8.
Blood ; 129(14): 1901-1912, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28179275

RESUMEN

Granulocyte colony-stimulating factor (G-CSF) is used clinically to treat leukopenia and to enforce hematopoietic stem cell (HSC) mobilization to the peripheral blood (PB). However, G-CSF is also produced in response to infection, and excessive exposure reduces HSC repopulation capacity. Previous work has shown that dormant HSCs contain all the long-term repopulation potential in the bone marrow (BM), and that as HSCs accumulate a divisional history, they progressively lose regenerative potential. As G-CSF treatment also induces HSC proliferation, we sought to examine whether G-CSF-mediated repopulation defects are a result of increased proliferative history. To do so, we used an established H2BGFP label retaining system to track HSC divisions in response to G-CSF. Our results show that dormant HSCs are preferentially mobilized to the PB on G-CSF treatment. We find that this mobilization does not result in H2BGFP label dilution of dormant HSCs, suggesting that G-CSF does not stimulate dormant HSC proliferation. Instead, we find that proliferation within the HSC compartment is restricted to CD41-expressing cells that function with short-term, and primarily myeloid, regenerative potential. Finally, we show CD41 expression is up-regulated within the BM HSC compartment in response to G-CSF treatment. This emergent CD41Hi HSC fraction demonstrates no observable engraftment potential, but directly matures into megakaryocytes when placed in culture. Together, our results demonstrate that dormant HSCs mobilize in response to G-CSF treatment without dividing, and that G-CSF-mediated proliferation is restricted to cells with limited regenerative potential found within the HSC compartment.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/farmacología , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/metabolismo , Glicoproteína IIb de Membrana Plaquetaria/biosíntesis , Animales , Proliferación Celular/genética , Regulación de la Expresión Génica/genética , Células Madre Hematopoyéticas/citología , Ratones , Ratones Transgénicos , Glicoproteína IIb de Membrana Plaquetaria/genética
9.
Blood ; 128(9): 1282-9, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27235135

RESUMEN

In addition to mutations in ITG2B or ITGB3 genes that cause defective αIIbß3 expression and/or function in Glanzmann's thrombasthenia patients, platelet dysfunction can be a result of genetic variability in proteins that mediate inside-out activation of αIIbß3 The RASGRP2 gene is strongly expressed in platelets and neutrophils, where its encoded protein CalDAG-GEFI facilitates the activation of Rap1 and subsequent activation of integrins. We used next-generation sequencing (NGS) and whole-exome sequencing (WES) to identify 2 novel function-disrupting mutations in RASGRP2 that account for bleeding diathesis and platelet dysfunction in 2 unrelated families. By using a panel of 71 genes, we identified a homozygous change (c.1142C>T) in exon 10 of RASGRP2 in a 9-year-old child of Chinese origin (family 1). This variant led to a p.Ser381Phe substitution in the CDC25 catalytic domain of CalDAG-GEFI. In 2 Spanish siblings from family 2, WES identified a nonsense homozygous variation (c.337C>T) (p.Arg113X) in exon 5 of RASGRP2 CalDAG-GEFI expression was markedly reduced in platelets from all patients, and by using a novel in vitro assay, we found that the nucleotide exchange activity was dramatically reduced in CalDAG-GEFI p.Ser381Phe. Platelets from homozygous patients exhibited agonist-specific defects in αIIbß3 integrin activation and aggregation. In contrast, α- and δ-granule secretion, platelet spreading, and clot retraction were not markedly affected. Integrin activation in the patients' neutrophils was also impaired. These patients are the first cases of a CalDAG-GEFI deficiency due to homozygous RASGRP2 mutations that are linked to defects in both leukocyte and platelet integrin activation.


Asunto(s)
Plaquetas/metabolismo , Exones , Factores de Intercambio de Guanina Nucleótido , Mutación Missense , Activación Plaquetaria/genética , Trombastenia , Proteínas de Unión al GTP rap1/metabolismo , Sustitución de Aminoácidos , Plaquetas/patología , Niño , Activación Enzimática/genética , Femenino , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Integrina beta3/genética , Integrina beta3/metabolismo , Masculino , Persona de Mediana Edad , Glicoproteína IIb de Membrana Plaquetaria/genética , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Vesículas Secretoras/genética , Vesículas Secretoras/metabolismo , Trombastenia/genética , Trombastenia/metabolismo , Trombastenia/patología
10.
Blood ; 125(24): 3769-77, 2015 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-25908104

RESUMEN

Mice with a constitutive or platelet-specific deletion of the C-type-lectin-like receptor (CLEC-2) exhibit hemorrhaging in the brain at mid-gestation. We sought to investigate the basis of this defect, hypothesizing that it is mediated by the loss of CLEC-2 activation by its endogenous ligand, podoplanin, which is expressed on the developing neural tube. To induce deletion of podoplanin at the 2-cell stage, we generated a podoplanin(fl/fl) mouse crossed to a PGK-Cre mouse. Using 3-dimensional light-sheet microscopy, we observed cerebral vessels were tortuous and aberrantly patterned at embryonic (E) day 10.5 in podoplanin- and CLEC-2-deficient mice, preceding the formation of large hemorrhages throughout the fore-, mid-, and hindbrain by E11.5. Immunofluorescence and electron microscopy revealed defective pericyte recruitment and misconnections between the endothelium of developing blood vessels and surrounding pericytes and neuro-epithelial cells. Nestin-Cre-driven deletion of podoplanin on neural progenitors also caused widespread cerebral hemorrhaging. Hemorrhaging was also seen in the ventricles of embryos deficient in the platelet integrin subunit glycoprotein IIb or in embryos in which platelet α-granule and dense granule secretion is abolished. We propose a novel role for podoplanin on the neuro-epithelium, which interacts with CLEC-2 on platelets, mediating platelet adhesion, aggregation, and secretion to guide the maturation and integrity of the developing vasculature and prevent hemorrhage.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/embriología , Circulación Cerebrovascular , Lectinas Tipo C/genética , Glicoproteínas de Membrana/genética , Animales , Plaquetas/metabolismo , Tipificación del Cuerpo , Encéfalo/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Hemorragias Intracraneales/genética , Hemorragias Intracraneales/metabolismo , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos C57BL , Activación Plaquetaria , Agregación Plaquetaria , Glicoproteína IIb de Membrana Plaquetaria/genética , Glicoproteína IIb de Membrana Plaquetaria/metabolismo
11.
Platelets ; 28(1): 82-89, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27534900

RESUMEN

Autophagy is an effective strategy for cell development by recycling cytoplasmic constituents. Genetic deletion of autophagy mediator Atg7 in hematopoietic stem cells (HSCs) can lead to failure of megakaryopoiesis and enhanced autophagy has been implicated in various hematological disorders such as immune thrombocytopenia and myelodysplastic syndrome. Here, we examined the hypothesis that optimal autophagy is essential for megakaryopoiesis and thrombopoiesis by altering autophagy using pharmacological approaches. When autophagy was induced by rapamycin or inhibited by bafilomycin A1 in fetal liver cells, we observed a significant decrease in high ploidy megakaryocytes, a reduction of CD41 and CD61 co-expressing cells, and less proplatelet or platelet formation. Additionally, reduced cell size was shown in megakaryocytes derived from rapamycin, but not bafilomycin A1-treated mouse fetal liver cells. However, when autophagy was altered in mature megakaryocytes, we observed no significant change in proplatelet formation, which was consistent with normal platelet counts, megakaryocyte numbers, and ploidy in Atg7flox/flox PF4-Cre mice with megakaryocyte- and platelet-specific deletion of autophagy-related gene Atg7. Therefore, our findings suggest that either induction or inhibition of autophagy in the early stage of megakaryopoiesis suppresses megakaryopoiesis and thrombopoiesis.


Asunto(s)
Autofagia/efectos de los fármacos , Macrólidos/farmacología , Sirolimus/farmacología , Trombopoyesis/efectos de los fármacos , Animales , Biomarcadores , Plaquetas/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Femenino , Citometría de Flujo , Expresión Génica , Integrina beta3/genética , Integrina beta3/metabolismo , Megacariocitos/citología , Megacariocitos/efectos de los fármacos , Megacariocitos/metabolismo , Ratones , Ratones Transgénicos , Recuento de Plaquetas , Glicoproteína IIb de Membrana Plaquetaria/genética , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Ploidias , Trombopoyesis/genética
12.
Indian J Med Res ; 145(5): 629-634, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28948953

RESUMEN

BACKGROUND & OBJECTIVES: Glanzmann thrombasthenia (GT) is a rare, inherited autosomal recessive disorder characterized by qualitative or quantitative deficiency of integrin αIIbß3 [glycoprotein IIb (GPIIb)/IIIa, CD41/CD61] diagnosed by absent or reduced platelet aggregation to physiological agonists, namely, collagen, adenosine-di-phosphate, epinephrine and arachidonic acid. The objective of this study was to quantitate platelet surface GPs, classify GT patients and relate the results with the severity of bleeding and platelet aggregation studies. METHODS: Fifty one patients of GT diagnosed by platelet aggregation studies were evaluated for the expression of CD41, CD61, CD42a and CD42b on platelet surface by flow cytometry. The association between the clinical phenotype based on bleeding score and GT subtype on flow cytometric evaluation was assessed. RESULTS: Twenty four (47%) patients of GT were classified as type I (as CD41/CD61 were virtually absent, <5%), six (11.8%) patients as type II (5-20% CD41/CD61) and 21 (41.2%) as type III or GT variants as they had near normal levels of CD41 and CD61. Type III GT patients had significantly lower numbers of severe bleeders (P=0.034), but the severity of bleeding did not vary significantly in type I and II GT patients. In all GT patients, mean CD41 expression was found to be lower than mean CD61 expression (P=0.002). INTERPRETATION & CONCLUSIONS: Type I GT was found most common in our patients and with lowered mean CD41 expression in comparison with CD61. Type III GT patients had significantly lower numbers of severe bleeders, but the severity of bleeding did not vary significantly in type I and II GT patients.


Asunto(s)
Hemorragia/sangre , Integrina beta3/genética , Glicoproteína IIb de Membrana Plaquetaria/genética , Trombastenia/genética , Adulto , Plaquetas/metabolismo , Plaquetas/patología , Femenino , Citometría de Flujo , Regulación de la Expresión Génica/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Glicoproteínas/sangre , Hemorragia/genética , Hemorragia/patología , Humanos , Integrina beta3/sangre , Masculino , Persona de Mediana Edad , Fenotipo , Agregación Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Glicoproteína IIb de Membrana Plaquetaria/sangre , Trombastenia/sangre , Trombastenia/patología
13.
Blood ; 124(24): 3587-96, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25298035

RESUMEN

Ecotropic viral integration site 1 (Evi1) is a transcription factor that is highly expressed in hematopoietic stem cells and is crucial for their self-renewal capacity. Aberrant expression of Evi1 is observed in 5% to 10% of de novo acute myeloid leukemia (AML) patients and predicts poor prognosis, reflecting multiple leukemogenic properties of Evi1. Here, we show that thrombopoietin (THPO) signaling is implicated in growth and survival of Evi1-expressing cells using a mouse model of Evi1 leukemia. We first identified that the expression of megakaryocytic surface molecules such as ITGA2B (CD41) and the THPO receptor, MPL, positively correlates with EVI1 expression in AML patients. In agreement with this finding, a subpopulation of bone marrow and spleen cells derived from Evi1 leukemia mice expressed both CD41 and Mpl. CD41(+) Evi1 leukemia cells induced secondary leukemia more efficiently than CD41(-) cells in a serial bone marrow transplantation assay. Importantly, the CD41(+) cells predominantly expressing Mpl effectively proliferated and survived on OP9 stromal cells in the presence of THPO via upregulating BCL-xL expression, suggesting an essential role of the THPO/MPL/BCL-xL cascade in enhancing the progression of Evi1 leukemia. These observations provide a novel aspect of the diverse functions of Evi1 in leukemogenesis.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Leucemia/metabolismo , Neoplasias Experimentales/metabolismo , Proteínas Oncogénicas/metabolismo , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Receptores de Trombopoyetina/metabolismo , Transducción de Señal , Trombopoyetina/metabolismo , Factores de Transcripción/metabolismo , Animales , Supervivencia Celular , Proteínas de Unión al ADN/genética , Regulación Leucémica de la Expresión Génica/genética , Leucemia/genética , Leucemia/patología , Proteína del Locus del Complejo MDS1 y EV11 , Ratones , Trasplante de Neoplasias , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Proteínas Oncogénicas/genética , Glicoproteína IIb de Membrana Plaquetaria/genética , Proto-Oncogenes/genética , Receptores de Trombopoyetina/genética , Trombopoyetina/genética , Factores de Transcripción/genética , Células Tumorales Cultivadas , Proteína bcl-X/biosíntesis , Proteína bcl-X/genética
14.
Tumour Biol ; 37(3): 3765-74, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26468019

RESUMEN

The initiation and formation of haemangioblastoma (HB) neovascularisation remain unknown, with concomitant controversy on its cytological origin. We detected HB-derived specific haematopoietic progenitors identified by surface expression of CD41 and CD45, which are similar to human embryonic vasculogenesis. CD41/CD45 cells expressed mesodermal markers, including SCL, Flk1 and c-kit. CD41 also seemed to appear before CD45 on haematopoietic progenitors. In vitro analysis showed that the CD41(+)/CD45 subpopulation gave rise to occasional primitive erythroid activity and endothelial marker expression. Meanwhile, kinetic investigation of the CD41(+)/CD45(+) subpopulation showed that some molecules, including SCL, Flk1 and c-kit, were involved in vascular formation. The CD45(+)/c-kit(+) population that lacked primitive haematopoiesis came from CD41(+) cells. Acquisition of CD45 expression by the haematopoietic progenitors was associated with advanced differentiation towards the vascular cell lineage. Taken together, the present data suggested that CD41 and CD45 expression marked the onset of HB neovascularisation and the stepwise development of the angioformative period. Our findings provide new insights into the mechanisms of HB neovascularisation and the underlying therapeutic targets of anti-vascular treatment.


Asunto(s)
Neoplasias Cerebelosas/metabolismo , Hemangioblastoma/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Neovascularización Patológica/metabolismo , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Adolescente , Adulto , Anciano , Diferenciación Celular/genética , Línea Celular , Línea Celular Tumoral , Neoplasias Cerebelosas/irrigación sanguínea , Neoplasias Cerebelosas/genética , Femenino , Hemangioblastoma/irrigación sanguínea , Hemangioblastoma/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Immunoblotting , Inmunohistoquímica , Antígenos Comunes de Leucocito/genética , Masculino , Microscopía Confocal , Persona de Mediana Edad , Neovascularización Patológica/genética , Glicoproteína IIb de Membrana Plaquetaria/genética , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Adulto Joven
15.
Transfusion ; 56(9): 2286-95, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27443848

RESUMEN

BACKGROUND: Pathogen inactivation (PI) techniques use ultraviolet (UV) illumination with or without a photosensitizer to destroy pathogen RNA and DNA. Although lacking a nucleus and innate DNA transcription, platelets (PLTs) contain RNA and can synthesize proteins. The impact of PI on PLT protein synthesis and function is unknown; altered synthesis may affect overall PLT quality. In this study we determine to what extent PLT RNA is affected by PI. STUDY DESIGN AND METHODS: In a pool-and-split design, paired apheresis PLT concentrates were treated with riboflavin and UV illumination or were left untreated. PLT total RNA and mRNA amounts specific for glycoproteins (GP)IIIa, GPIIb, and GPIb; α-granule proteins PLT factor (PF)4; osteonectin and thrombospondin (TSP); and housekeeping protein glyceraldehyde-3-phosphate dehydrogenase (GAPDH) were determined using absorbance and quantitative polymerase chain reaction. RESULTS: After treatment, amounts of all analyzed mRNAs were significantly reduced (p < 0.05), but to different degrees. For GAPDH and PF4, transcripts appeared less susceptible to the treatment, with 70% remaining 1 hour after UV illumination. For GPIIIa and TSP, less than 15% remained after treatment. There was a correlation (R(2) = 0.85) between transcript length and amount of mRNA remaining 1 hour after treatment. Total RNA demonstrated a life span equal to the PLT life span of 10 to 11 days. CONCLUSION: This is the first report of the impact of riboflavin and UV illumination on PLT mRNA. Results suggest that all mRNA present in PLTs is affected by the treatment although the degree of the effect varies among transcripts.


Asunto(s)
Plaquetas/metabolismo , ARN Mensajero/genética , Riboflavina/farmacología , Rayos Ultravioleta , Plaquetas/efectos de los fármacos , Plaquetas/efectos de la radiación , Conservación de la Sangre/métodos , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/análisis , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/genética , Humanos , Integrina beta3/análisis , Integrina beta3/genética , Osteonectina/análisis , Osteonectina/genética , Factor Plaquetario 4/análisis , Factor Plaquetario 4/genética , Glicoproteína IIb de Membrana Plaquetaria/análisis , Glicoproteína IIb de Membrana Plaquetaria/genética , ARN Mensajero/efectos de los fármacos , ARN Mensajero/efectos de la radiación , Trombospondinas/análisis , Trombospondinas/genética
16.
Blood Cells Mol Dis ; 54(1): 78-83, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25135204

RESUMEN

Morpholino and vivo-morpholino gene knockdown methods have been used to study thrombocyte function in zebrafish. However, a large-scale knockdown of the entire zebrafish genome using these technologies to study thrombocyte function is prohibitively expensive. We have developed an inexpensive gene knockdown method, which uses a hybrid of a control vivo-morpholino and a standard antisense oligonucleotide specific for a gene. This hybrid molecule is able to deliver antisense deoxyoligonucleotides into zebrafish thrombocytes because it piggybacks on a control vivo-morpholino. To validate use of this hybrid molecule in gene knockdowns, we targeted the thrombocyte specific αIIb gene with a hybrid of a control vivo-morpholino and an oligonucleotide antisense to αIIb mRNA. The use of this piggyback technology resulted in degradation of αIIb mRNA and led to thrombocyte functional defect. This piggyback method to knockdown genes is inexpensive since one control vivo-morpholino can be used to target many different genes by making many independent gene-specific oligonucleotide hybrids. Thus, this novel piggyback technology can be utilized for cost-effective large-scale knockdowns of genes to study thrombocyte function in zebrafish.


Asunto(s)
Plaquetas/metabolismo , Técnicas de Silenciamiento del Gen/métodos , Morfolinos , Glicoproteína IIb de Membrana Plaquetaria , Estabilidad del ARN , Proteínas de Pez Cebra , Pez Cebra , Animales , Morfolinos/genética , Morfolinos/farmacología , Glicoproteína IIb de Membrana Plaquetaria/genética , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Estabilidad del ARN/efectos de los fármacos , Estabilidad del ARN/genética , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
17.
Blood ; 121(5): 770-80, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23169780

RESUMEN

UNLABELLED: Several studies have demonstrated that hematopoietic cells originate from endotheliumin early development; however, the phenotypic progression of progenitor cells during human embryonic hemogenesis is not well described. Here, we define the developmental hierarchy among intermediate populations of hematopoietic progenitor cells (HPCs) derived from human embryonic stem cells (hESCs). We genetically modified hESCs to specifically demarcate acquisition of vascular (VE-cadherin) and hematopoietic (CD41a) cell fate and used this dual-reporting transgenic hESC line to observe endothelial to hematopoietic transition by real-time confocal microscopy. Live imaging and clonal analyses revealed a temporal bias in commitment of HPCs that recapitulates discrete waves of lineage differentiation noted during mammalian hemogenesis. Specifically, HPCs isolated at later time points showed reduced capacity to form erythroid/ megakaryocytic cells and exhibited a tendency toward myeloid fate that was enabled by expression of the Notch ligand Dll4 on hESC-derived vascular feeder cells. These data provide a framework for defining HPC lineage potential, elucidate a molecular contribution from the vascular niche in promoting hematopoietic lineage progression, and distinguish unique subpopulations of hemogenic endothelium during hESC differentiation. KEY POINTS: Live imaging of endothelial to hematopoietic conversion identifies distinct subpopulations of hESC-derived hemogenic endothelium. Expression of the Notch ligand DII4 on vascular ECs drives induction of myeloid fate from hESC-derived hematopoietic progenitors.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/metabolismo , Células Endoteliales/metabolismo , Células Madre Hematopoyéticas/metabolismo , Transducción Genética , Antígenos CD/biosíntesis , Antígenos CD/genética , Cadherinas/biosíntesis , Cadherinas/genética , Técnicas de Cocultivo , Células Madre Embrionarias/citología , Células Endoteliales/citología , Células Nutrientes , Células Madre Hematopoyéticas/citología , Humanos , Glicoproteína IIb de Membrana Plaquetaria/biosíntesis , Glicoproteína IIb de Membrana Plaquetaria/genética
18.
Blood ; 121(22): 4463-72, 2013 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-23564910

RESUMEN

The hematopoietic stem cell (HSC) compartment is heterogeneous, yet our understanding of the identities of different HSC subtypes is limited. Here we show that platelet integrin CD41 (αIIb), currently thought to only transiently mark fetal HSCs, is expressed on an adult HSC subtype that accumulates with age. CD41+ HSCs were largely quiescent and exhibited myeloerythroid and megakaryocyte gene priming, governed by Gata1, whereas CD41- HSCs were more proliferative and exhibited lymphoid gene priming. When isolated without the use of blocking antibodies, CD41+ HSCs possessed long-term repopulation capacity on serial transplantations and showed a marked myeloid bias compared with CD41- HSCs, which yielded a more lymphoid-biased progeny. CD41-knockout (KO) mice displayed multilineage hematopoietic defects coupled with decreased quiescence and survival of HSCs, suggesting that CD41 is functionally relevant for HSC maintenance and hematopoietic homeostasis. Transplantation experiments indicated that CD41-KO-associated defects are long-term transplantable, HSC-derived and, in part, mediated through the loss of platelet mass leading to decreases in HSC exposure to important platelet released cytokines, such as transforming growth factor ß1. In summary, our data provide a novel marker to identify a myeloid-biased HSC subtype that becomes prevalent with age and highlights the dogma of HSC regulation by their progeny.


Asunto(s)
Células Madre Adultas/metabolismo , Envejecimiento/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Progenitoras Mieloides/metabolismo , Glicoproteína IIb de Membrana Plaquetaria/genética , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Células Madre Adultas/citología , Envejecimiento/inmunología , Animales , Biomarcadores/metabolismo , Plaquetas/citología , Plaquetas/metabolismo , Linaje de la Célula/inmunología , Supervivencia Celular/inmunología , Citometría de Flujo , Expresión Génica/inmunología , Hematopoyesis/inmunología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Ratones , Ratones Noqueados , Células Progenitoras Mieloides/citología , Factor de Crecimiento Transformador beta/metabolismo
19.
Platelets ; 26(8): 779-82, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25734216

RESUMEN

Glanzmann's thrombasthenia (GT) is an inherited disorder of platelet aggregation, characterized by qualitative and quantitative defect on platelet αIIbß3 integrin (GpIIb/IIIa), resulting in lifelong bleeding tendency due to defective platelet plug formation. The αIIb gene (ITGA2B) and ß3 gene (ITGB3) are closely located at chromosome 17q21.31-32. ITGA2B consist of 30 exons and encoding α chain, whereas ITGB3 has 15 exons and encoding ß chain. Until now, according to the Human Gene Mutation Database (HGMD), 138 mutations at ITGA2B gene and 101 mutations at ITGB3 gene have been identified. We aimed to determine whether there was any mutation in the ITGA2B and ITGB3 genes, and a correlation between clinical phenotype and genotype in Turkish GT patients. We examined 20 patients with GT followed at the Department of Pediatric Hematology, Meram Faculty of Medicine, for Clinical and Laboratory Findings and Molecular Genetic Analysis. Peripheral blood was collected from patients, and a written informed consent for genetic analysis was obtained from parents. DNA was isolated from by proteinase K and phenol/chloroform extraction. ITGA2B and ITGB3 genes were screened by polymerase chain reaction. There were 12 females and 8 males with a median age of 15.25 years. Major clinical presentations of these patients were mucocutaneous bleedings. The most common bleeding type was epistaxis (85%). Life-threatening bleedings were seen in five patients. Seven (35%) patients showed various mutations in the ITGA2B or ITGB3 genes. We detected four novel mutations in three different regions and two mutations defined previously within the ITGA2B gene. These changes are at exon 4; c.570 T > G alteration, at exon 13 c.1277 T > A, c.1291 T > G alterations, at exon 19 c.1921A > G alterations. And from the start point of exon 14, behind 107 bases, we detected a heterozygous alteration at Thymine to Guanine. According to PolyPhen Database Program and NCBI Multiple Alignment Tool Database, four transitions are conserved at evolutionary process, so we can say that these transitions are novel mutations. c. 468T > G alteration at exon 4 and c. 1378 T > A alteration at exon 13 were reported to HGMD previously. Screening the exons of the ITGB3 gene from the same patient groups, we reported a novel missense mutation at exon 5, at nucleotide 680. No correlation was found between clinical phenotype and genotype. These mutations were described for the first time in Turkish population, and all novel mutations are not defined previously. Furthermore, collaborative studies are needed for the population point of view.


Asunto(s)
Integrina beta3/genética , Mutación , Glicoproteína IIb de Membrana Plaquetaria/genética , Trombastenia/genética , Adolescente , Adulto , Plaquetas/metabolismo , Niño , Preescolar , Consanguinidad , Exones , Femenino , Genotipo , Humanos , Masculino , Fenotipo , Agregación Plaquetaria , Recuento de Plaquetas , Pruebas de Función Plaquetaria , Trombastenia/diagnóstico , Turquía , Adulto Joven
20.
Proc Natl Acad Sci U S A ; 108(29): 11890-5, 2011 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-21730166

RESUMEN

Integrins are large cell-surface adhesion receptors that can be activated to a high affinity state by the formation of an intracellular complex between the integrin ß-subunit tail, the membrane, and talin. The F2 and F3 subdomains of the talin head play a key role in formation of this complex. Here, activation of the integrin αIIb/ß3 dimer by the talin head domain was probed using multiscale molecular dynamics simulations. A number of novel insights emerge from these studies, including (i) the importance of the integrin αIIb subunit F992 and F993 residues in stabilizing the "off" state of the αIIb/ß3 dimer, (ii) a crucial role for negatively charged groups in the F2-F3/membrane interaction, (iii) binding of the talin F2-F3 domain to negatively charged lipid headgroups in the membrane induces a reorientation of the ß transmembrane (TM) domain, (iv) an increase in the tilt angle of the ß TM domain relative to the bilayer normal helps to destabilize the α/ß TM interaction and promote a scissor-like movement of the integrin TM helices. These results, combined with various published experimental observations, suggest a model for the mechanism of inside-out activation of integrins by talin.


Asunto(s)
Membrana Celular/metabolismo , Integrina beta3/metabolismo , Modelos Moleculares , Complejos Multiproteicos/metabolismo , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Talina/metabolismo , Dimerización , Integrina beta3/genética , Simulación de Dinámica Molecular , Mutación/genética , Glicoproteína IIb de Membrana Plaquetaria/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA