Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Transfusion ; 62(1): 173-182, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34757639

RESUMEN

BACKGROUND: The COVID19 pandemic highlights the need for contingency planning in the event of blood shortages. To increase platelet supply, we assessed the operational impact and effect on platelet quality of splitting units prior to storage. STUDY DESIGN AND METHODS: Using production figures, we modeled the impact on unit numbers, platelet counts, and volumes of splitting only apheresis double donations into three units (yielding ⅔ doses), or all standard dose units in half. To assess quality, eight pools of three ABO/Rh-matched apheresis (Trima Accel) double donations in plasma were split to ⅔ and ½ volumes in both Terumo and Fresenius storage bags. These were irradiated and subject to maximal permitted periods of nonagitation (3 × 8 h) before comparing platelet quality markers (including pH, CD62P expression) to Day 9 of storage. RESULTS: Splitting all double donations into three predicted inventory expansion of 23% overall whereas halving all standard dose units clearly doubles stock. In our study, ⅔ and ½ doses contained 153 ± 15 × 109 (~138 ml) and 113 ± 11 × 109 (~102 ml) platelets respectively. Following storage, higher pH was observed in ⅔ than in ½ doses and in Terumo compared to Fresenius bags. The higher pH was reflected in better quality markers, including lower CD62P expression. Despite the differences, on Day 8 (of pH monitoring at expiry) all ⅔ doses and most ½ doses were ≥pH 6.4. CONCLUSION: A strategy to split apheresis platelets in plasma to lower doses is feasible, maintains acceptable platelet quality, and should be considered by blood services in response to extreme shortages.


Asunto(s)
Plaquetas , COVID-19 , Plaquetas/metabolismo , Conservación de la Sangre , Humanos , Recuento de Plaquetas , Plaquetoferesis
2.
Blood ; 131(9): 1000-1011, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29187380

RESUMEN

Mutations in NBEAL2, the gene encoding the scaffolding protein Nbeal2, are causal of gray platelet syndrome (GPS), a rare recessive bleeding disorder characterized by platelets lacking α-granules and progressive marrow fibrosis. We present here the interactome of Nbeal2 with additional validation by reverse immunoprecipitation of Dock7, Sec16a, and Vac14 as interactors of Nbeal2. We show that GPS-causing mutations in its BEACH domain have profound and possible effects on the interaction with Dock7 and Vac14, respectively. Proximity ligation assays show that these 2 proteins are physically proximal to Nbeal2 in human megakaryocytes. In addition, we demonstrate that Nbeal2 is primarily localized in the cytoplasm and Dock7 on the membrane of or in α-granules. Interestingly, platelets from GPS cases and Nbeal2-/- mice are almost devoid of Dock7, resulting in a profound dysregulation of its signaling pathway, leading to defective actin polymerization, platelet activation, and shape change. This study shows for the first time proteins interacting with Nbeal2 and points to the dysregulation of the canonical signaling pathway of Dock7 as a possible cause of the aberrant formation of platelets in GPS cases and Nbeal2-deficient mice.


Asunto(s)
Plaquetas/metabolismo , Proteínas Sanguíneas/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Megacariocitos/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Plaquetas/citología , Proteínas Sanguíneas/genética , Proteínas Activadoras de GTPasa/genética , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Megacariocitos/citología , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Mutación , Unión Proteica , Proteínas de Transporte Vesicular/genética
3.
Transfusion ; 59(9): 2952-2963, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31294868

RESUMEN

BACKGROUND: Rejuvenation of stored red blood cells (RBCs) increases levels of adenosine 5'-triphosphate (ATP) and 2,3-diphosphoglycerate (2,3-DPG) to those of fresh cells. This study aimed to optimize and validate the US-approved process to a UK setting for manufacture and issue of rejuvenated RBCs for a multicenter randomized controlled clinical trial in cardiac surgery. STUDY DESIGN AND METHODS: Rejuvenation of leukoreduced RBC units involved adding a solution containing pyruvate, inosine, phosphate, and adenine (Rejuvesol, Zimmer Biomet), warming at 37°C for 60 minutes, then "manual" washing with saline adenine glucose mannitol solution. A laboratory study was conducted on six pools of ABO/D-matched units made the day after donation. On Days 7, 21, and 28 of 4 ± 2°C storage, one unit per pool was rejuvenated and measured over 96 hours for volume, hematocrit, hemolysis, ATP, 2,3-DPG, supernatant potassium, lactate, and purines added (inosine) or produced (hypoxanthine) by rejuvenation. Subsequently, an operational validation (two phases of 32 units each) was undertaken, with results from the first informing a trial component specification applied to the second. Rejuvenation effects were also tested on crossmatch reactivity and RBC antigen profiles. RESULTS: Rejuvenation raised 2,3-DPG to, and ATP above, levels of fresh cells. The final component had potassium and hemolysis values below those of standard storage Days 7 and 21, respectively, containing 1.2% exogenous inosine and 500 to 1900 µmoles/unit of hypoxanthine. The second operational validation met compliance to the trial component specification. Rejuvenation did not adversely affect crossmatch reactivity or RBC antigen profiles. CONCLUSION: The validated rejuvenation process operates within defined quality limits, preserving RBC immunophenotypes, enabling manufacture for clinical trials.


Asunto(s)
Conservación de la Sangre/métodos , Eritrocitos/fisiología , Medicina Regenerativa/métodos , Rejuvenecimiento/fisiología , 2,3-Difosfoglicerato/metabolismo , Adenosina Trifosfato/sangre , Tipificación y Pruebas Cruzadas Sanguíneas , Pérdida de Sangre Quirúrgica/prevención & control , Conservación de la Sangre/normas , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Criopreservación/métodos , Recuento de Eritrocitos , Transfusión de Eritrocitos/normas , Eritrocitos/citología , Hemólisis/fisiología , Humanos , Inmunofenotipificación , Materiales Manufacturados , Purinas/sangre , Control de Calidad , Ensayos Clínicos Controlados Aleatorios como Asunto , Medicina Regenerativa/normas
5.
Br J Haematol ; 175(3): 381-392, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27650431

RESUMEN

Allogeneic platelet transfusions are widely used for the prevention and treatment of bleeding in thrombocytopenia. Recent evidence suggests platelet transfusions have limited efficacy and are associated with uncertain immunomodulatory risks and concerns about viral or bacterial transmission. Alternatives to transfusion are a well-recognised tenet of Patient Blood Management, but there has been less focus on different strategies to reduce bleeding risk by comparison to platelet transfusion. Direct alternatives to platelet transfusion include agents to stimulate endogenous platelet production (thrombopoietin mimetics), optimising platelet adhesion to endothelium by treating anaemia or increasing von Willebrand factor levels (desmopressin), increasing formation of cross-linked fibrinogen (activated recombinant factor VII, fibrinogen concentrate or recombinant factor XIII), decreasing fibrinolysis (tranexamic acid or epsilon aminocaproic acid) or using artificial or modified platelets (cryopreserved platelets, lyophilised platelets, haemostatic particles, liposomes, engineered nanoparticles or infusible platelet membranes). The evidence base to support the use of these alternatives is variable, but an area of active research. Much of the current randomised controlled trial focus is on evaluation of the use of thrombopoietin mimetics and anti-fibrinolytics. It is also recognised that one alternative strategy to platelet transfusion is choosing not to transfuse at all.


Asunto(s)
Terapias Complementarias , Hemorragia/prevención & control , Hemorragia/terapia , Transfusión de Plaquetas , Trombocitopenia/complicaciones , Antifibrinolíticos/uso terapéutico , Mimetismo Biológico , Factores de Coagulación Sanguínea/uso terapéutico , Transfusión Sanguínea , Terapias Complementarias/métodos , Humanos , Nanopartículas , Transfusión de Plaquetas/efectos adversos , Trombopoyetina/uso terapéutico
6.
Platelets ; 27(6): 526-34, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27135140

RESUMEN

A goal of platelet storage is to maintain the quality of platelets from the point of donation to the point of transfusion - to suspend the aging process. This effort is judged by clinical and laboratory measures with varying degrees of success. Recent work gives encouragement that platelets can be maintained ex vivo beyond the current 5 -7 day shelf life whilst maintaining their quality, as measured by posttransfusion recovery and survival. However, additional measures are needed to validate the development of technologies that may further reduce the aging of stored platelets, or enhance their hemostatic properties.


Asunto(s)
Plaquetas/fisiología , Conservación de la Sangre , Senescencia Celular , Transfusión de Plaquetas , Animales , Conservación de la Sangre/efectos adversos , Seguridad de la Sangre , Humanos , Transfusión de Plaquetas/efectos adversos , Transfusión de Plaquetas/métodos , Transfusión de Plaquetas/normas , Plasma Rico en Plaquetas , Resultado del Tratamiento
7.
Blood ; 120(24): 4859-68, 2012 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-22972982

RESUMEN

We recently identified 68 genomic loci where common sequence variants are associated with platelet count and volume. Platelets are formed in the bone marrow by megakaryocytes, which are derived from hematopoietic stem cells by a process mainly controlled by transcription factors. The homeobox transcription factor MEIS1 is uniquely transcribed in megakaryocytes and not in the other lineage-committed blood cells. By ChIP-seq, we show that 5 of the 68 loci pinpoint a MEIS1 binding event within a group of 252 MK-overexpressed genes. In one such locus in DNM3, regulating platelet volume, the MEIS1 binding site falls within a region acting as an alternative promoter that is solely used in megakaryocytes, where allelic variation dictates different levels of a shorter transcript. The importance of dynamin activity to the latter stages of thrombopoiesis was confirmed by the observation that the inhibitor Dynasore reduced murine proplatelet for-mation in vitro.


Asunto(s)
Plaquetas/metabolismo , Dinamina III/genética , Genoma Humano/genética , Proteínas de Homeodominio/genética , Megacariocitos/metabolismo , Proteínas de Neoplasias/genética , Regiones Promotoras Genéticas/genética , Animales , Sitios de Unión/genética , Plaquetas/efectos de los fármacos , Línea Celular Tumoral , Linaje de la Célula/genética , Células Cultivadas , Inmunoprecipitación de Cromatina , Expresión Génica , Variación Genética , Proteínas de Homeodominio/metabolismo , Humanos , Hidrazonas/farmacología , Ratones , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteínas de Neoplasias/metabolismo , Recuento de Plaquetas , Polimorfismo de Nucleótido Simple , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Sitio de Iniciación de la Transcripción , Transcripción Genética
8.
Transfusion ; 54(6): 1478-85, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24308361

RESUMEN

BACKGROUND: Studies show that 1 in 1200 neonates have a low platelet (PLT) count due to alloimmunization against human PLT antigen (HPA)-1a (ß3 -L33). This mainly occurs in HPA-1a-negative mothers who are positive for the human leukocyte antigen (HLA)-DRB3*01:01 allele, but only about one-third of cases will mount an effective alloimmune response. The development of specific treatment modalities requires that the mechanisms driving the maternal alloimmune response against the fetal PLTs be further explored. An antibody reagent that has a different binding affinity to HLA-DRA/DRB3*01:01 with and without the ß3 -L33 peptide would be a valuable reagent to study peptide presentation on maternal antigen-presenting cells. STUDY DESIGN AND METHODS: To identify such antibodies, HLA-DRA/DRB3*01:01 was recombinantly expressed in Drosophila S2 cells. To delineate the epitope of interesting antibodies, seven mutant HLA-DRA/DRB3*01:01 molecules were generated by site-directed mutagenesis introducing naturally occurring amino acid changes encoded by DRB3*02 and DRB3*03 alleles. RESULTS: The murine monoclonal antibody (MoAb) DA2 showed robust binding by enzyme-linked immunosorbent assay to recombinant HLA-DRA/DRB3*01:01, but binding was reduced in the presence of ß3 -L33 peptide. The binding affinity of DA2 to the mutant HLA-DRA/DRB3*0101 in which serine at Position 60 of the ß1-chain was replaced by tyrosine was greatly enhanced. Interestingly the binding of DA2 to the mutant was not reduced by the presence of ß3 -L33 peptide. CONCLUSION: The results of this study generate a molecular model of the interaction of the HLA-DRA/DRB3*01:01 molecule with MoAb DA2. This will inform functional studies with the recombinant Class II molecules.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Antígenos HLA/metabolismo , Cadenas alfa de HLA-DR/metabolismo , Cadenas HLA-DRB3/metabolismo , Antígenos de Plaqueta Humana/metabolismo , Sitios de Unión , Ensayo de Inmunoadsorción Enzimática , Cadenas alfa de HLA-DR/química , Cadenas HLA-DRB3/química , Humanos , Integrina beta3 , Unión Proteica , Estructura Secundaria de Proteína
9.
Blood ; 115(24): 5069-79, 2010 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-20351310

RESUMEN

Exposed subendothelial collagen acts as a substrate for platelet adhesion and thrombus formation after vascular injury. Synthetic collagen-derived triple-helical peptides, designated collagen-related peptide (CRP), GFOGER, and VWF-III, can specifically engage the platelet collagen receptors, glycoprotein VI and integrin alpha(2)beta(1), and plasma von Willebrand factor (VWF), respectively. Hitherto, the role of these 3 collagen-binding axes has been studied indirectly. Use of these uniform peptide substrates, rather than collagen fibers, provides independent control of each axis. Here, we use confocal imaging and novel image analysis techniques to investigate the effects of receptor-ligand engagement on platelet binding and activation during thrombus formation under flow conditions. At low shear (100s(-1) and 300s(-1)), both GFOGER and CRP are required for thrombus formation. At 1000s(-1), a combination of either CRP or GFOGER with VWF-III induces comparable thrombus formation, and VWF-III increases thrombus deposition at all shear rates, being indispensable at 3000s(-1). A combination of CRP and VWF-III is sufficient to support extensive platelet deposition at 3000s(-1), with slight additional effect of GFOGER. Measurement of thrombus height after specific receptor blockade or use of altered proportions of peptides indicates a signaling rather than adhesive role for glycoprotein VI, and primarily adhesive roles for both alpha(2)beta(1) and the VWF axis.


Asunto(s)
Plaquetas/metabolismo , Colágeno/metabolismo , Integrina alfa2beta1/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores de Colágeno/metabolismo , Trombosis/metabolismo , Proteínas Portadoras/metabolismo , Humanos , Imitación Molecular , Fragmentos de Péptidos/metabolismo , Péptidos/metabolismo , Adhesividad Plaquetaria/fisiología , Unión Proteica/fisiología , Flujo Sanguíneo Regional/fisiología , Transducción de Señal/fisiología , Estrés Mecánico , Factor de von Willebrand/metabolismo
10.
Blood Adv ; 6(18): 5415-5428, 2022 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-35736672

RESUMEN

Stored red blood cells (RBCs) incur biochemical and morphological changes, collectively termed the storage lesion. Functionally, the storage lesion manifests as slower oxygen unloading from RBCs, which may compromise the efficacy of transfusions where the clinical imperative is to rapidly boost oxygen delivery to tissues. Recent analysis of large real-world data linked longer storage with increased recipient mortality. Biochemical rejuvenation with a formulation of adenosine, inosine, and pyruvate can restore gas-handling properties, but its implementation is impractical for most clinical scenarios. We tested whether storage under hypoxia, previously shown to slow biochemical degradation, also preserves gas-handling properties of RBCs. A microfluidic chamber, designed to rapidly switch between oxygenated and anoxic superfusates, was used for single-cell oxygen saturation imaging on samples stored for up to 49 days. Aliquots were also analyzed flow cytometrically for side-scatter (a proposed proxy of O2 unloading kinetics), metabolomics, lipidomics, and redox proteomics. For benchmarking, units were biochemically rejuvenated at 4 weeks of standard storage. Hypoxic storage hastened O2 unloading in units stored to 35 days, an effect that correlated with side-scatter but was not linked to posttranslational modifications of hemoglobin. Although hypoxic storage and rejuvenation produced distinct biochemical changes, a subset of metabolites including pyruvate, sedoheptulose 1-phosphate, and 2/3 phospho-d-glycerate, was a common signature that correlated with changes in O2 unloading. Correlations between gas handling and lipidomic changes were modest. Thus, hypoxic storage of RBCs preserves key metabolic pathways and O2 exchange properties, thereby improving the functional quality of blood products and potentially influencing transfusion outcomes.


Asunto(s)
Conservación de la Sangre , Oxígeno , Adenosina/metabolismo , Conservación de la Sangre/métodos , Eritrocitos/metabolismo , Hemoglobinas/metabolismo , Humanos , Hipoxia/metabolismo , Inosina/metabolismo , Oxígeno/metabolismo , Fosfatos/metabolismo , Piruvatos/metabolismo
11.
Blood ; 111(10): 4986-96, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18305222

RESUMEN

We have analyzed the adhesion of human and murine platelets, and of recombinant human and murine GpVI ectodomains, to synthetic triple-helical collagen-like peptides. These included 57 peptides derived from the sequence of human type III collagen and 9 peptides derived from the cyanogen bromide fragment of bovine type III collagen, alpha1(III)CB4. We have identified several peptides that interact with GpVI, in particular a peptide designated III-30 with the sequence GAOGLRGGAGPOGPEGGKGAAGPOGPO. Both human and murine platelets bound to peptide III-30 in a GpVI-dependent manner. III-30 also supported binding of recombinant GpVI ectodomains. Cross-linked III-30 induced aggregation of human and murine platelets, although with a lower potency than collagen-related peptide. Modifications of the peptide sequence indicated that the hydroxyproline residues play a significant role in supporting its GpVI reactivity. However, many peptides containing OGP/GPO motifs did not support adhesion to GpVI. These data indicate that the ability of a triple-helical peptide to bind GpVI is not solely determined by the presence or spatial arrangement of these OGP/GPO motifs within the peptides.


Asunto(s)
Colágeno Tipo III/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Plaquetas/metabolismo , Bovinos , Colágeno Tipo III/química , Humanos , Ratones , Fragmentos de Péptidos , Péptidos/síntesis química , Péptidos/química , Agregación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/química , Unión Proteica
12.
J Immunol Methods ; 316(1-2): 75-83, 2006 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-17027020

RESUMEN

We report the development of an expression system for the production of soluble, calmodulin (CaM)-tagged proteins in Drosophila Schneider S2 cells and the subsequent use of these proteins for the selection of phage displayed antibodies. The CaM-tag permitted the purification of recombinant protein to >90% purity in a single step at yields of >20 mg/l. Using platelet glycoprotein VI (GP6) as a model, we demonstrated that the recombinant CaM-tagged protein was post-translationally N-glycosylated and had identical ligand specificity to native protein. A novel selection strategy, exploiting the CaM tag, was then used to isolate four single chain Fv fragments (scFvs) specific for GP6 from a non-immune phage display library. In contrast to other selection methods, which can result in antibodies that do not recognise native protein, all of the scFvs we selected bound cell surface expressed GP6. In conclusion, the production of CaM-tagged proteins in Drosophila Schneider S2 cells and the selection strategy reported here offer advantages over previously published methods, including simple culture conditions, rapid protein purification, specific elution of phage antibodies and preferential selection of phage antibodies that recognise native, cell surface expressed protein.


Asunto(s)
Antígenos/biosíntesis , Calmodulina/genética , Drosophila melanogaster/genética , Fragmentos de Inmunoglobulinas/biosíntesis , Biblioteca de Péptidos , Glicoproteínas de Membrana Plaquetaria/genética , Proteínas Recombinantes/biosíntesis , Secuencia de Aminoácidos , Animales , Antígenos/genética , Western Blotting , Calmodulina/metabolismo , Línea Celular , Clonación Molecular , Drosophila melanogaster/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Fragmentos de Inmunoglobulinas/genética , Fragmentos de Inmunoglobulinas/inmunología , Datos de Secuencia Molecular , Glicoproteínas de Membrana Plaquetaria/inmunología , Proteínas Recombinantes/síntesis química , Proteínas Recombinantes/genética
13.
FASEB J ; 19(8): 898-909, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15923400

RESUMEN

Lipid-rich atherosclerotic plaques are vulnerable, and their rupture can cause the formation of a platelet- and fibrin-rich thrombus leading to myocardial infarction and ischemic stroke. Although the role of plaque-based tissue factor as stimulator of blood coagulation has been recognized, it is not known whether plaques can cause thrombus formation through direct activation of platelets. We isolated lipid-rich atheromatous plaques from 60 patients with carotid stenosis and identified morphologically diverse collagen type I- and type III-positive structures in the plaques that directly stimulated adhesion, dense granule secretion, and aggregation of platelets in buffer, plasma, and blood. This material also elicited platelet-monocyte aggregation and platelet-dependent blood coagulation. Plaques exposed to flowing blood at arterial wall shear rate induced platelets to adhere to and spread on the collagenous structures, triggering subsequent thrombus formation. Plaque-induced platelet thrombus formation was observed in fully anticoagulated blood (i.e., in the absence of tissue factor-mediated coagulation). Mice platelets lacking glycoprotein VI (GPVI) were unable to adhere to atheromatous plaque or form thrombi. Human platelet thrombus formation onto plaques in flowing blood was completely blocked by GPVI inhibition with the antibody 10B12 but not affected by integrin alpha2beta1 inhibition with 6F1 mAb. Moreover, the initial platelet response, shape change, induced by plaque was blocked by GPVI inhibition but not with alpha2beta1 antagonists (6F1 mAb or GFOGER-GPP peptide). Pretreatment of plaques with collagenase or anti-collagen type I and anti-collagen type III antibodies abolished plaque-induced platelet activation. Our results indicate that morphologically diverse collagen type I- and collagen type III-containing structures in lipid-rich atherosclerotic plaques stimulate thrombus formation by activating platelet GPVI. This platelet collagen receptor, essential for plaque-induced thrombus formation, presents a promising new anti-thrombotic target for the prevention of ischemic cardiovascular diseases.


Asunto(s)
Aterosclerosis/complicaciones , Glicoproteínas de Membrana Plaquetaria/fisiología , Trombosis/etiología , Animales , Aterosclerosis/patología , Coagulación Sanguínea , Plaquetas/fisiología , Estenosis Carotídea/sangre , Colágeno Tipo I/análisis , Colágeno Tipo III/análisis , Humanos , Integrina alfa2beta1/fisiología , Lípidos/análisis , Ratones , Microscopía Fluorescente , Monocitos/fisiología , Activación Plaquetaria , Adhesividad Plaquetaria , Agregación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores
14.
Thromb Haemost ; 88(4): 648-54, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12362238

RESUMEN

Calreticulin is an abundant protein in the endoplasmic reticulum of most cells. In this study, flow cytometry and immunoprecipitation from surface-biotinylated platelets each provided direct evidence that calreticulin is also expressed on the surface of human platelets. Anti-calreticulin antibodies caused platelet activation, inducing FcgammaRIIa-independent platelet aggregation. In addition, these antibodies inhibited platelet adhesion to the integrin alpha2beta1-specific ligands, GFOGER-GPP and monomeric collagen I, and to the glycoprotein VI-specific ligand, CRP. Inhibition of platelet adhesion to these ligands was independent of integrin alphaIIbbeta3. In resting platelets, calreticulin was shown to interact with integrin alpha2beta1 and glycoprotein VI. Together, these data demonstrate that surface calreticulin is associated with collagen receptors on the platelet surface, where it may play a role in the modulation of the platelet-collagen interaction.


Asunto(s)
Plaquetas/metabolismo , Calreticulina/metabolismo , Membrana Celular/química , Integrina alfa2beta1/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Anticuerpos/farmacología , Plaquetas/química , Plaquetas/ultraestructura , Calreticulina/inmunología , Calreticulina/fisiología , Colágeno/farmacología , Humanos , Integrina alfa2beta1/química , Integrina alfa2beta1/fisiología , Ligandos , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria , Glicoproteínas de Membrana Plaquetaria/química , Glicoproteínas de Membrana Plaquetaria/fisiología , Receptores de Colágeno/metabolismo , Trombastenia/patología
15.
Thromb Haemost ; 91(4): 743-54, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15045136

RESUMEN

Recombinant HPA-1a antibodies with Fc, mutated to remove destructive effector functions, have been developed as a potential therapy for fetomaternal alloimmune thrombocytopenia (FMAIT), via blockade of binding of human HPA-1a polyclonal antibodies to fetal HPA-1a1b platelets. We have assessed the effect of the IgG1 HPA-1a antibody B2G1 and two mutated derivatives in various functional assays in resting and agonist-stimulated platelets of the three HPA-1 genotypes. With HPA-1a1b platelets (fetal genotype), the antibodies did not activate signalling or CD62P expression in resting platelets, did not change in vitro bleeding time (IVBT), and did not inhibit platelet adhesion to collagen in flowing blood. Adhesion of HPA-1a1b platelets to fibrinogen was reduced by 20%, and aggregation induced by ADP by 50%, but collagen-related peptide (CRP-XL)-induced aggregation was normal. With HPA-1a1a platelets, aggregation to both ADP and CRP-XL was inhibited, with total blockade of adhesion to fibrinogen and of IVBT responses. Interestingly, a monovalent antibody fragment with identical specificity had no inhibitory effect on aggregation. In static adhesion assays using human alphaIIbbeta3 or alphaVbeta3 transfectants of HPA-1a (Leu(33)) phenotype, attachment to fibrinogen of the latter but not of the former was completely blocked by the HPA-1a antibodies. These observations are best explained by antibody-mediated blockade of the RGD binding site on beta3 by a mechanism of steric hindrance. As the effect on platelet function is modest with HPA-1a1b (fetal type) platelets, the mutated HPA-1a antibodies described here could be developed further for FMAIT therapy.


Asunto(s)
Anticuerpos/farmacología , Antígenos de Plaqueta Humana/inmunología , Activación Plaquetaria/inmunología , Polimorfismo de Nucleótido Simple/inmunología , Anticuerpos/genética , Anticuerpos/inmunología , Colágeno/metabolismo , Femenino , Fibrinógeno/metabolismo , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/farmacología , Integrina alfaVbeta3/metabolismo , Integrina beta3/inmunología , Leucina , Pruebas de Función Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Embarazo , Ingeniería de Proteínas , Proteínas Recombinantes
16.
Nat Genet ; 45(5): 542-545, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23563608

RESUMEN

The blood group Vel was discovered 60 years ago, but the underlying gene is unknown. Individuals negative for the Vel antigen are rare and are required for the safe transfusion of patients with antibodies to Vel. To identify the responsible gene, we sequenced the exomes of five individuals negative for the Vel antigen and found that four were homozygous and one was heterozygous for a low-frequency 17-nucleotide frameshift deletion in the gene encoding the 78-amino-acid transmembrane protein SMIM1. A follow-up study showing that 59 of 64 Vel-negative individuals were homozygous for the same deletion and expression of the Vel antigen on SMIM1-transfected cells confirm SMIM1 as the gene underlying the Vel blood group. An expression quantitative trait locus (eQTL), the common SNP rs1175550 contributes to variable expression of the Vel antigen (P = 0.003) and influences the mean hemoglobin concentration of red blood cells (RBCs; P = 8.6 × 10(-15)). In vivo, zebrafish with smim1 knockdown showed a mild reduction in the number of RBCs, identifying SMIM1 as a new regulator of RBC formation. Our findings are of immediate relevance, as the homozygous presence of the deletion allows the unequivocal identification of Vel-negative blood donors.


Asunto(s)
Antígenos de Grupos Sanguíneos/genética , Membrana Eritrocítica/metabolismo , Eritrocitos/inmunología , Eliminación de Gen , Homocigoto , Proteínas de la Membrana/genética , Sitios de Carácter Cuantitativo , Alelos , Animales , Biomarcadores/metabolismo , Antígenos de Grupos Sanguíneos/inmunología , Antígenos de Grupos Sanguíneos/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Eritrocitos/metabolismo , Eritrocitos/patología , Exoma/genética , Femenino , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Isoanticuerpos/inmunología , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Embarazo , Pez Cebra/genética
17.
Nat Genet ; 44(4): 435-9, S1-2, 2012 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-22366785

RESUMEN

The exon-junction complex (EJC) performs essential RNA processing tasks. Here, we describe the first human disorder, thrombocytopenia with absent radii (TAR), caused by deficiency in one of the four EJC subunits. Compound inheritance of a rare null allele and one of two low-frequency SNPs in the regulatory regions of RBM8A, encoding the Y14 subunit of EJC, causes TAR. We found that this inheritance mechanism explained 53 of 55 cases (P < 5 × 10(-228)) of the rare congenital malformation syndrome. Of the 53 cases with this inheritance pattern, 51 carried a submicroscopic deletion of 1q21.1 that has previously been associated with TAR, and two carried a truncation or frameshift null mutation in RBM8A. We show that the two regulatory SNPs result in diminished RBM8A transcription in vitro and that Y14 expression is reduced in platelets from individuals with TAR. Our data implicate Y14 insufficiency and, presumably, an EJC defect as the cause of TAR syndrome.


Asunto(s)
Predisposición Genética a la Enfermedad , Proteínas de Unión al ARN/genética , Trombocitopenia/genética , Deformidades Congénitas de las Extremidades Superiores/genética , Regiones no Traducidas 5'/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Niño , Preescolar , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Femenino , Variación Genética , Humanos , Lactante , Recién Nacido , Masculino , Mutación , Recuento de Plaquetas , Polimorfismo de Nucleótido Simple , Radio (Anatomía)/anomalías , Alineación de Secuencia , Análisis de Secuencia de ADN , Trombocitopenia/congénito , Adulto Joven , Pez Cebra/genética
18.
Nat Genet ; 43(8): 735-7, 2011 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-21765411

RESUMEN

Gray platelet syndrome (GPS) is a predominantly recessive platelet disorder that is characterized by mild thrombocytopenia with large platelets and a paucity of α-granules; these abnormalities cause mostly moderate but in rare cases severe bleeding. We sequenced the exomes of four unrelated individuals and identified NBEAL2 as the causative gene; it has no previously known function but is a member of a gene family that is involved in granule development. Silencing of nbeal2 in zebrafish abrogated thrombocyte formation.


Asunto(s)
Plaquetas/metabolismo , Gránulos Citoplasmáticos/metabolismo , Síndrome de Plaquetas Grises/genética , Proteínas del Tejido Nervioso/genética , Vesículas Secretoras/metabolismo , Adulto , Anciano , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Plaquetas/patología , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Linaje , Análisis de Secuencia de ADN , Homología de Secuencia de Ácido Nucleico , Adulto Joven , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
19.
Dev Cell ; 20(5): 597-609, 2011 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21571218

RESUMEN

Hematopoietic differentiation critically depends on combinations of transcriptional regulators controlling the development of individual lineages. Here, we report the genome-wide binding sites for the five key hematopoietic transcription factors--GATA1, GATA2, RUNX1, FLI1, and TAL1/SCL--in primary human megakaryocytes. Statistical analysis of the 17,263 regions bound by at least one factor demonstrated that simultaneous binding by all five factors was the most enriched pattern and often occurred near known hematopoietic regulators. Eight genes not previously appreciated to function in hematopoiesis that were bound by all five factors were shown to be essential for thrombocyte and/or erythroid development in zebrafish. Moreover, one of these genes encoding the PDZK1IP1 protein shared transcriptional enhancer elements with the blood stem cell regulator TAL1/SCL. Multifactor ChIP-Seq analysis in primary human cells coupled with a high-throughput in vivo perturbation screen therefore offers a powerful strategy to identify essential regulators of complex mammalian differentiation processes.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Factor de Transcripción GATA1/metabolismo , Factor de Transcripción GATA2/metabolismo , Genoma Humano/genética , Megacariocitos/metabolismo , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Sitios de Unión , Diferenciación Celular , Células Cultivadas , Humanos , Megacariocitos/citología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína 1 de la Leucemia Linfocítica T Aguda
20.
J Biol Chem ; 282(2): 1296-304, 2007 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-17085439

RESUMEN

Collagen-related peptide is a selective agonist for the platelet collagen receptor Glycoprotein VI. The triple helical peptide contains ten GPO triplets/strand (single letter amino acid nomenclature, where O is hydroxyproline) and so over-represents GPO compared with native collagen sequence. To investigate the ability of Glycoprotein VI to recognize GPO triplets in a setting more representative of the collagens, we synthesized a set of triple helical peptides containing fewer GPO triplets, varying their number and spacing within an inert (GPP)n backbone. The adhesion of recombinant human Glycoprotein VI ectodo-main, like that of human platelets, to these peptides increased with their GPO content, and platelet adhesion was abolished by the specific anti-Glycoprotein VI-blocking antibody, 10B12. Platelet aggregation and protein tyrosine phosphorylation were induced only by cross-linked peptides and only those that contained two or more GPO triplets. Such peptides were less potent than cross-linked collagen-related peptide. Our data suggest that both the sequences GPOGPO and GPO.........GPO represent functional Glycoprotein VI recognition motifs within collagen. Furthermore, we propose that the (GPO)4 motif can support simultaneous binding of two glycoprotein VI molecules, in either a parallel or anti-parallel stacking arrangement, which could play an important role in activation of signaling.


Asunto(s)
Plaquetas/metabolismo , Colágeno/química , Colágeno/metabolismo , Adhesividad Plaquetaria/fisiología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Sitios de Unión/fisiología , Reactivos de Enlaces Cruzados/química , Reactivos de Enlaces Cruzados/metabolismo , Glicina/metabolismo , Humanos , Hidroxiprolina/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fosforilación , Agregación Plaquetaria/fisiología , Glicoproteínas de Membrana Plaquetaria/química , Prolina/metabolismo , Unión Proteica/fisiología , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal/fisiología , Relación Estructura-Actividad , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA