Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Angiogenesis ; 24(3): 677-693, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33770321

RESUMEN

Endothelial barrier disruption and vascular leak importantly contribute to organ dysfunction and mortality during inflammatory conditions like sepsis and acute respiratory distress syndrome. We identified the kinase Arg/Abl2 as a mediator of endothelial barrier disruption, but the role of Arg in endothelial monolayer regulation and its relevance in vivo remain poorly understood. Here we show that depletion of Arg in endothelial cells results in the activation of both RhoA and Rac1, increased cell spreading and elongation, redistribution of integrin-dependent cell-matrix adhesions to the cell periphery, and improved adhesion to the extracellular matrix. We further show that Arg is activated in the endothelium during inflammation, both in murine lungs exposed to barrier-disruptive agents, and in pulmonary microvessels of septic patients. Importantly, Arg-depleted endothelial cells were less sensitive to barrier-disruptive agents. Despite the formation of F-actin stress fibers and myosin light chain phosphorylation, Arg depletion diminished adherens junction disruption and intercellular gap formation, by reducing the disassembly of cell-matrix adhesions and cell retraction. In vivo, genetic deletion of Arg diminished vascular leak in the skin and lungs, in the presence of a normal immune response. Together, our data indicate that Arg is a central and non-redundant regulator of endothelial barrier integrity, which contributes to cell retraction and gap formation by increasing the dynamics of adherens junctions and cell-matrix adhesions in a Rho GTPase-dependent fashion. Therapeutic inhibition of Arg may provide a suitable strategy for the treatment of a variety of clinical conditions characterized by vascular leak.


Asunto(s)
Matriz Extracelular/metabolismo , Uniones Comunicantes/enzimología , Células Endoteliales de la Vena Umbilical Humana/enzimología , Proteínas Tirosina Quinasas/metabolismo , Alveolos Pulmonares/enzimología , Animales , Adhesión Celular/genética , Activación Enzimática , Matriz Extracelular/genética , Uniones Comunicantes/genética , Humanos , Inflamación/enzimología , Inflamación/genética , Ratones , Ratones Noqueados , Proteínas Tirosina Quinasas/genética
2.
Angiogenesis ; 24(3): 695-714, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33983539

RESUMEN

Sprouting angiogenesis is key to many pathophysiological conditions, and is strongly regulated by vascular endothelial growth factor (VEGF) signaling through VEGF receptor 2 (VEGFR2). Here we report that the early endosomal GTPase Rab5C and its activator RIN2 prevent lysosomal routing and degradation of VEGF-bound, internalized VEGFR2 in human endothelial cells. Stabilization of endosomal VEGFR2 levels by RIN2/Rab5C is crucial for VEGF signaling through the ERK and PI3-K pathways, the expression of immediate VEGF target genes, as well as specification of angiogenic 'tip' and 'stalk' cell phenotypes and cell sprouting. Using overexpression of Rab mutants, knockdown and CRISPR/Cas9-mediated gene editing, and live-cell imaging in zebrafish, we further show that endosomal stabilization of VEGFR2 levels is required for developmental angiogenesis in vivo. In contrast, the premature degradation of internalized VEGFR2 disrupts VEGF signaling, gene expression, and tip cell formation and migration. Thus, an endosomal feedforward mechanism maintains receptor signaling by preventing lysosomal degradation, which is directly linked to the induction of target genes and cell fate in collectively migrating cells during morphogenesis.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica , Proteolisis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Animales , Proteínas Portadoras/genética , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Pez Cebra/genética , Proteínas de Unión al GTP rab5/genética
3.
Hum Genomics ; 14(1): 39, 2020 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-33066815

RESUMEN

The expression of the human ß-like globin genes follows a well-orchestrated developmental pattern, undergoing two essential switches, the first one during the first weeks of gestation (ε to γ), and the second one during the perinatal period (γ to ß). The γ- to ß-globin gene switching mechanism includes suppression of fetal (γ-globin, HbF) and activation of adult (ß-globin, HbA) globin gene transcription. In hereditary persistence of fetal hemoglobin (HPFH), the γ-globin suppression mechanism is impaired leaving these individuals with unusual elevated levels of fetal hemoglobin (HbF) in adulthood. Recently, the transcription factors KLF1 and BCL11A have been established as master regulators of the γ- to ß-globin switch. Previously, a genomic variant in the KLF1 gene, identified by linkage analysis performed on twenty-seven members of a Maltese family, was found to be associated with HPFH. However, variation in the levels of HbF among family members, and those from other reported families carrying genetic variants in KLF1, suggests additional contributors to globin switching. ASF1B was downregulated in the family members with HPFH. Here, we investigate the role of ASF1B in γ- to ß-globin switching and erythropoiesis in vivo. Mouse-human interspecies ASF1B protein identity is 91.6%. By means of knockdown functional assays in human primary erythroid cultures and analysis of the erythroid lineage in Asf1b knockout mice, we provide evidence that ASF1B is a novel contributor to steady-state erythroid differentiation, and while its loss affects the balance of globin expression, it has no major role in hemoglobin switching.


Asunto(s)
Proteínas de Ciclo Celular/genética , Eritropoyesis/genética , Chaperonas de Histonas/genética , Globinas beta/genética , Animales , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Regulación de la Expresión Génica , Células HEK293 , Chaperonas de Histonas/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones Noqueados , Polimorfismo de Nucleótido Simple , Interferencia de ARN , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , gamma-Globinas/genética
4.
Platelets ; 30(3): 368-379, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29553857

RESUMEN

Platelet concentrates (PCs) represent a blood transfusion product with a major concern for safety as their storage temperature (20-24°C) allows bacterial growth, and their maximum storage time period (less than a week) precludes complete microbiological testing. Pathogen inactivation technologies (PITs) provide an additional layer of safety to the blood transfusion products from known and unknown pathogens such as bacteria, viruses, and parasites. In this context, PITs, such as Mirasol Pathogen Reduction Technology (PRT), have been developed and are implemented in many countries. However, several studies have shown in vitro that Mirasol PRT induces a certain level of platelet shape change, hyperactivation, basal degranulation, and increased oxidative damage during storage. It has been suggested that Mirasol PRT might accelerate what has been described as the platelet storage lesion (PSL), but supportive molecular signatures have not been obtained. We aimed at dissecting the influence of both variables, that is, Mirasol PRT and storage time, at the proteome level. We present comprehensive proteomics data analysis of Control PCs and PCs treated with Mirasol PRT at storage days 1, 2, 6, and 8. Our workflow was set to perform proteomics analysis using a gel-free and label-free quantification (LFQ) approach. Semi-quantification was based on LFQ signal intensities of identified proteins using MaxQuant/Perseus software platform. Data are available via ProteomeXchange with identifier PXD008119. We identified marginal differences between Mirasol PRT and Control PCs during storage. However, those significant changes at the proteome level were specifically related to the functional aspects previously described to affect platelets upon Mirasol PRT. In addition, the effect of Mirasol PRT on the platelet proteome appeared not to be exclusively due to an accelerated or enhanced PSL. In summary, semi-quantitative proteomics allows to discern between proteome changes due to Mirasol PRT or PSL, and proves to be a methodology suitable to phenotype platelets in an unbiased manner, in various physiological contexts.


Asunto(s)
Plaquetas/metabolismo , Pruebas de Función Plaquetaria/métodos , Proteoma/metabolismo , Proteómica/métodos , Humanos
5.
J Immunol ; 197(11): 4312-4324, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27815426

RESUMEN

Dendritic cells (DCs) play a pivotal role in the regulation of the immune response. DC development and activation is finely orchestrated through transcriptional programs. GATA1 transcription factor is required for murine DC development, and data suggest that it might be involved in the fine-tuning of the life span and function of activated DCs. We generated DC-specific Gata1 knockout mice (Gata1-KODC), which presented a 20% reduction of splenic DCs, partially explained by enhanced apoptosis. RNA sequencing analysis revealed a number of deregulated genes involved in cell survival, migration, and function. DC migration toward peripheral lymph nodes was impaired in Gata1-KODC mice. Migration assays performed in vitro showed that this defect was selective for CCL21, but not CCL19. Interestingly, we show that Gata1-KODC DCs have reduced polysialic acid levels on their surface, which is a known determinant for the proper migration of DCs toward CCL21.


Asunto(s)
Movimiento Celular/inmunología , Quimiocina CCL21/inmunología , Células Dendríticas/inmunología , Factor de Transcripción GATA1/inmunología , Ganglios Linfáticos/inmunología , Ácidos Siálicos/inmunología , Animales , Movimiento Celular/genética , Quimiocina CCL19/genética , Quimiocina CCL19/inmunología , Quimiocina CCL21/genética , Células Dendríticas/citología , Factor de Transcripción GATA1/deficiencia , Ganglios Linfáticos/citología , Ratones , Ratones Noqueados , Ácidos Siálicos/genética
7.
Blood Cells Mol Dis ; 55(4): 293-303, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26460250

RESUMEN

Dendritic cells (DCs) are key initiators and regulators of the immune response. The development of the DC lineage and their subsets requires an orchestrated regulation of their transcriptional program. Gata1, a transcription factor expressed in several hematopoietic cell lineages, has been recently reported to be required for mouse DC development and function. In humans, GATA1 is involved in the lineage separation between monocyte-derived DCs and Langerhans cells (LC) and loss of GATA1 results in differentiation arrest at the monocyte stage. The hematopoietic GATA factors (i.e. Gata1, Gata2, Gata3) are known to regulate each other's expression and to function consecutively throughout lineage commitment (so-called GATA switch). In humans, mutations in GATA2 are causative of MonoMAC disease, a human immunodeficiency syndrome characterized by loss of DCs, monocytes, B and NK cells. However, additional data on the expression of hematopoietic GATA factors in the DC lineage is missing. In this study, we have characterized the expression of hematopoietic GATA factors in murine and human DCs and their expression dynamics upon TLR stimulation. We found that all hematopoietic GATA factors are expressed in DCs, but identified species-specific differences in the relative expression of each GATA factor, and how their expression fluctuates upon stimulation.


Asunto(s)
Células Dendríticas/metabolismo , Factores de Transcripción GATA/genética , Regulación de la Expresión Génica , Hematopoyesis/genética , Empalme Alternativo , Animales , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Factores de Transcripción GATA/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-4/metabolismo , Interleucina-4/farmacología , Ratones , Monocitos/metabolismo , Isoformas de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
Blood ; 121(10): e70-80, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23303822

RESUMEN

The main function of platelets is to maintain normal hemostasis. Inefficient platelet production and/or defective platelet function results in bleeding disorders resulting from a wide range of genetic traits and acquired pathologies. Several platelet function tests have been developed for use in the clinic and in experimental animal models. In particular, platelet aggregation is routinely measured in an aggregometer, which requires normal platelet counts and significant blood sample volumes. For this reason, the analysis of thrombocytopenic patients, infants, and animal models is problematic. We have developed a novel flow cytometry test of platelet aggregation, in which 10- to 25-fold lower platelet counts or sample volumes can be used, either of platelet-rich plasma or whole blood from human subjects or mice. This setup can be applied to test in small assay volumes the influence of a variety of stimuli, drugs, and plasma factors, such as antibodies, on platelet aggregation. The presented principle stands as a novel promising tool, which allows analysis of platelet aggregation in thrombocytopenic patients or infants, and facilitates studies in platelets obtained from experimental animal models without the need of special devices but a flow cytometer.


Asunto(s)
Plaquetas/metabolismo , Citometría de Flujo/métodos , Síndrome de Deficiencia de Adhesión del Leucocito/sangre , Agregación Plaquetaria , Trombastenia/sangre , Animales , Autoanticuerpos/análisis , Autoanticuerpos/sangre , Plaquetas/patología , Estudios de Casos y Controles , Humanos , Síndrome de Deficiencia de Adhesión del Leucocito/diagnóstico , Ratones , Activación Plaquetaria , Recuento de Plaquetas , Plasma Rico en Plaquetas , Trombastenia/diagnóstico
9.
Blood ; 119(2): 583-6, 2012 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-22065596

RESUMEN

Patients with Glanzmann thrombasthenia or Leukocyte Adhesion Deficiency-III syndrome (LAD-III or LAD-1/variant) present with increased bleeding tendency because of the lack or dysfunction of the fibrinogen receptor GPIIb/IIIa (integrin αIIbß3), respectively. Although the bleeding disorder is more severe in LAD-III patients, classic aggregometry or perfusion of Glanzmann or LAD-III platelets over collagen-coated slides under physiologic shear rate does not discriminate between these 2 conditions. However, in a novel flow cytometry-based aggregation assay, Glanzmann platelets were still capable of forming small aggregates upon collagen stimulation, whereas LAD-III platelets were not. These aggregates required functional GPIa/IIa (integrin α2ß1) instead of integrin αIIbß3, thus explaining the clinically more severe bleeding manifestations in LAD-III patients, in which all platelet integrins are functionally defective. These findings provide genetic evidence for the differential requirements of platelet integrins in thrombus formation and demonstrate that correct integrin function assessment can be achieved with a combination of diagnostic methods.


Asunto(s)
Hemorragia/diagnóstico , Integrina alfa2beta1/metabolismo , Síndrome de Deficiencia de Adhesión del Leucocito/metabolismo , Adhesividad Plaquetaria/fisiología , Agregación Plaquetaria/fisiología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Trombastenia/metabolismo , Colágeno/metabolismo , Citometría de Flujo , Hemorragia/etiología , Hemorragia/metabolismo , Humanos , Síndrome de Deficiencia de Adhesión del Leucocito/complicaciones , Síndrome de Deficiencia de Adhesión del Leucocito/patología , Fenotipo , Trombastenia/complicaciones , Trombastenia/patología
10.
Transfusion ; 54(9): 2292-300, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24689812

RESUMEN

BACKGROUND: Recent studies showed that Mirasol pathogen reduction treatment (PRT) leads to increased P-selectin expression and increased oxygen and glucose consumption in resting platelets (PLTs). This study investigates the effect of PRT on PLT activation. STUDY DESIGN AND METHODS: Untreated or Mirasol-treated PLTs were analyzed at different time points during storage. Microaggregation upon stimulation with phorbol myristate acetate (PMA), convulxin, and ristocetin was measured. Alpha granule contents and release upon thrombin stimulation were assessed by flow cytometry and Western blotting. PLT spreading was determined on collagen-coated glass slides. RESULTS: Mirasol PRT led to spontaneous aggregation (hyperreactivity), as measured by flow cytometry in the absence of agonist throughout storage time. PMA-induced aggregation was significantly higher in Mirasol PRT PLTs compared to controls. Aggregation in response to convulxin and ristocetin was significantly lower and directly influenced by storage time after Mirasol PRT, compared to untreated stored PLT concentrates. Despite the reported hyperreactivity of resting PLTs, PLT activation with thrombin on Day 8 after Mirasol PRT resulted in less P-selectin-positive PLTs. Furthermore, platelet factor 4 (PF4) secretion was reduced upon thrombin stimulation on Day 8 after PRT compared to controls. Significantly decreased spreading of Mirasol PRT PLTs over collagen-coated slides was observed directly after PRT and persisted throughout storage. CONCLUSION: Mirasol PRT leads to hyperreactive PLTs, probably caused by continuous basal degranulation through storage time. This results in a reduction in the degranulation capacity upon acute stimulation, which influences PLT spreading, but not overtly microaggregation. The clinical relevance needs to be investigated.


Asunto(s)
Plaquetas/efectos de los fármacos , Plaquetas/efectos de la radiación , Riboflavina/farmacología , Rayos Ultravioleta , Plaquetas/metabolismo , Conservación de la Sangre/métodos , Colágeno/metabolismo , Citometría de Flujo , Humanos , Activación Plaquetaria/efectos de los fármacos , Activación Plaquetaria/efectos de la radiación , Transfusión de Plaquetas
12.
Blood Adv ; 6(11): 3529-3534, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35298626

RESUMEN

Immune thrombocytopenia (ITP) is an autoimmune disease characterized by enhanced platelet clearance and defective platelet production. Diagnosis by exclusion and trial-and-error treatment strategies is common practice, and despite the advancement in treatment options, many patients remain refractory. Although the existence of different pathophysiological entities is acknowledged, we are still far from stratifying and understanding ITP. To investigate, we sought to dissect the platelet proteome dynamics in so-called passive and active preclinical ITP mouse models, with which we propose to phenocopy respectively acute/newly diagnosed and persistent/chronic stages of ITP in humans. We obtained the platelet proteome at the thrombocytopenic stage and after platelet count recovery (reached naturally or by IVIg-treatment, depending on the model). Although most of the proteomic alterations were common to both ITP models, there were model-specific protein dynamics that accompanied and explained alterations in platelet aggregation responses, as measured in the passive ITP model. The expression dynamics observed in Syk may explain, extrapolated to humans and pending validation, the increased bleeding tendency of patients with ITP when treated with fostamatinib as third or later- as opposed to second line of treatment. We propose that the platelet proteome may give diagnostic and prognostic insights into ITP and that such studies should be pursued in humans.


Asunto(s)
Púrpura Trombocitopénica Idiopática , Trombocitopenia , Animales , Plaquetas , Modelos Animales de Enfermedad , Humanos , Ratones , Proteoma , Proteómica , Púrpura Trombocitopénica Idiopática/tratamiento farmacológico
13.
Blood Adv ; 5(23): 5116-5127, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34551092

RESUMEN

von Willebrand factor (VWF) is an essential hemostatic protein that is synthesized and secreted by endothelial cells and stored in Weibel-Palade bodies (WPBs). The secretory Rab GTPases Rab27A, Rab3B, and Rab3D have been linked with WPB trafficking and secretion. How these Rabs are activated and recruited to WPBs remains elusive. In this study, we identified MAP kinase-activating death domain (MADD) as the guanine nucleotide exchange factor for Rab27A and both Rab3 isoforms in primary human endothelial cells. Rab activity assays revealed a reduction in Rab27A, Rab3B, and Rab3D activation upon MADD silencing. Rab activation, but not binding, was dependent on the differentially expressed in normal and neoplastic cells (DENN) domain of MADD, indicating the potential existence of 2 Rab interaction modules. Furthermore, immunofluorescent analysis showed that Rab27A, Rab3B, and Rab3D recruitment to WPBs was dramatically decreased upon MADD knockdown, revealing that MADD drives Rab membrane targeting. Artificial mistargeting of MADD using a TOMM70 tag abolished Rab27A localization to WPB membranes in a DENN domain-dependent manner, indicating that normal MADD localization in the cytosol is crucial. Activation of Rab3B and Rab3D was reduced upon Rab27A silencing, suggesting that activation of these Rabs is enhanced through previous activation of Rab27A by MADD. MADD silencing did not affect WPB morphology, but it did reduce VWF intracellular content. Furthermore, MADD-depleted cells exhibited decreased histamine-evoked VWF release, similar to Rab27A-depleted cells. In conclusion, MADD acts as a master regulator of VWF secretion by coordinating the activation and membrane targeting of secretory Rabs to WPBs.


Asunto(s)
Cuerpos de Weibel-Palade , Proteínas de Unión al GTP rab , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte , Células Endoteliales/metabolismo , Exocitosis , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Guanosina Trifosfato , Humanos , Cuerpos de Weibel-Palade/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
14.
Blood ; 112(13): 4935-9, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18796633

RESUMEN

UV-C irradiation has been shown to be effective for pathogen reduction in platelet concentrates, but preliminary work indicated that UV-C irradiation of platelets can induce platelet aggregation. In this study, the mechanism underlying this phenomenon was investigated. Irradiation of platelets with UV-C light (1500 J/m(2)) caused platelet aggregation, which was dependent on integrin alphaIIbbeta3 activation (GPIIb/IIIa). This activation occurred despite treatment with several signal transduction inhibitors known to block platelet activation. UV-C also induced activation of recombinant alphaIIbbeta3 in Chinese hamster ovary (CHO) cells, an environment in which physiologic agonists fail to activate. Activation of alphaIIbbeta3 requires talin binding to the beta3 tail, yet alphaIIbbeta3-Delta724 (lacking the talin binding site) was activated by UV-C irradiation, excluding a requirement for talin binding. The UV-C effect appears to be general in that beta(1) and beta(2) integrins are also activated by UV-C. To explain these findings, we investigated the possibility of UV-C-induced photolysis of disulfide bonds, in analogy with the activating effect of reducing agents on integrins. Indeed, UV-C induced a marked increase in free thiol groups in platelet surface proteins including alphaIIbbeta3. Thus, UV-C appears to activate alphaIIbbeta3 not by affecting intracellular signal transduction, but by reduction of disulfide bonds regulating integrin conformation.


Asunto(s)
Plaquetas/química , Disulfuros/efectos de la radiación , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/efectos de la radiación , Rayos Ultravioleta/efectos adversos , Animales , Células CHO , Línea Celular Tumoral , Células Cultivadas , Cricetinae , Cricetulus , Humanos , Neutrófilos , Fotólisis , Agregación Plaquetaria/efectos de la radiación , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/efectos de la radiación , Conformación Proteica
15.
J Leukoc Biol ; 2020 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-33289106

RESUMEN

Dendritic cells (DCs) are key immune modulators and are able to mount immune responses or tolerance. DC differentiation and activation imply a plethora of molecular and cellular responses, including transcriptional changes. PU.1 is a highly expressed transcription factor in DCs and coordinates relevant aspects of DC biology. Due to their role as immune regulators, DCs pose as a promising immunotherapy tool. However, some of their functional features, such as survival, activation, or migration, are compromised due to the limitations to simulate in vitro the physiologic DC differentiation process. A better knowledge of transcriptional programs would allow the identification of potential targets for manipulation with the aim of obtaining "qualified" DCs for immunotherapy purposes. Most of the current knowledge regarding DC biology derives from studies using mouse models, which not always find a parallel in human. In the present study, we dissect the PU.1 transcriptional regulome and interactome in mouse and human DCs, in the steady state or LPS activated. The PU.1 transcriptional regulome was identified by performing PU.1 chromatin immunoprecipitation followed by high-throughput sequencing and pairing these data with RNAsequencing data. The PU.1 interactome was identified by performing PU.1 immunoprecipitation followed by mass spectrometry analysis. Our results portray PU.1 as a pivotal factor that plays an important role in the regulation of genes required for proper DC activation and function, and assures the repression of nonlineage genes. The interspecies differences between human and mouse DCs are surprisingly substantial, highlighting the need to study the biology of human DCs.

16.
Transfusion ; 49(6): 1233-41, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19290994

RESUMEN

BACKGROUND: Stored platelets (PLTs) are metabolically active, resulting in a decrease of pH during storage. The pH of PLT concentrates (PCs) is recognized as a measure of quality, and pH limits are set by regulatory bodies. A pH sensor was built into a PLT storage container, and the feasibility of testing pH using a noninvasive fluorescent measurement method was evaluated. STUDY DESIGN AND METHODS: A citrated polyvinylchloride (PVC) PLT storage container with pH sensor insert was made and evaluated for biocompatibility during PLT storage and on pH reading accuracy, reproducibility, and durability. A noninvasive fluorescence reader was tested versus syringe-based sampling and subsequent measurement with a blood gas analyzer (BGA). The effect of interfering substances in plasma on the accuracy of this optical measurement was tested. Calibration and accuracy of the pH sensor were determined in both phosphate-buffered saline and in PCs. RESULTS: The citrated PVC storage container with pH sensor insert showed good storage properties for 300 mL of pooled buffy coat PLTs in plasma over 7 days. The pH sensor was easy to use and tracked pH(22) in the range of 6.2 to 7.8 over 11 days of storage. Accuracy in PCs was 0.08 pH units measured at 22 degrees C when calibrated against a BGA. CONCLUSION: The storage container with integrated pH sensor and noninvasive reader allows pH of PCs to be tracked over time in a noninvasive manner.


Asunto(s)
Plaquetas/metabolismo , Tecnología de Fibra Óptica/instrumentación , Concentración de Iones de Hidrógeno , Calibración , Fluorometría , Humanos
17.
Thromb Res ; 184: 115-121, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31731068

RESUMEN

INTRODUCTION: Red blood cell (RBC) transfusion is associated with an increased risk of pro-thrombotic events, but the underlying mechanism is poorly understood. We hypothesized that RBC transfusion modulates platelet activity in critically ill patients with and without sepsis. METHODS: In a prospective cohort study, 37 critically ill patients receiving a single RBC unit to correct for anemia were sampled prior to and 1 h after transfusion. Platelet exposure of P-selectin, CD63 and binding of PAC-1 as well as formation of platelet-leukocyte complexes were measured by flow cytometry. The ability of plasma from critically ill patients to induce ex vivo platelet aggregation was assessed by flow cytometry after incubation with platelets from a healthy donor. RESULTS: RBC transfusion neither triggered the expression of platelet activation markers nor the formation of platelet-leukocyte complexes. Plasma from critically ill patients induced more spontaneous platelet aggregation prior to RBC transfusion compared to healthy controls, which was further augmented following RBC transfusion. Also collagen-induced platelet aggregation was already increased prior to RBC transfusion compared to healthy controls, and this response was unaffected by RBC transfusion. In contrast, ristocetin-induced platelet agglutination was decreased when compared to controls, suggesting impaired vWF-dependent platelet agglutination, even in the presence of high vWF levels. Following RBC transfusion, ristocetin-induced platelet agglutination further decreased. There were no differences between septic and non-septic recipients in all assays. CONCLUSION: Ex vivo platelet aggregation is disturbed in the critically ill. Transfusion of a RBC unit may further increase the spontaneous platelet aggregatory response.


Asunto(s)
Plaquetas/metabolismo , Transfusión de Eritrocitos/métodos , Activación Plaquetaria/fisiología , Anciano , Enfermedad Crítica , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos
18.
Antivir Ther ; 11(1): 105-16, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16518966

RESUMEN

In this study we investigated the long-term effect of highly active antiretroviral therapy (HAART) on HIV-specific CD4+ T-cell responses in comparison with virus-specific CD4+ T-cell responses against the persistent herpes viruses cytomegalovirus (CMV) and Epstein-Barr virus (EBV). To this end, HIV- and herpes virus-specific cellular immune responses were measured longitudinally in 10 seroconverters with long-term follow-up including 55 months of successful suppression of viral load by HAART. HIV- and CMV-specific CD4+ T cells producing interferon-gamma (IFNgamma) or interleukin-2 (IL-2) were analysed as well as proliferative capacity. EBV-specific CD4+ T cells were determined using a 12-day ex vivo assay. Initiation of HAART resulted in a transient increase of HIV-specific IL-2(+)IFNgamma(+)CD4(+) T cells and, to a lesser extent, IL-2(+)CD4(+) T cells. Long-term HAART resulted in an increase in HIV-, CMV- and EBV-specific CD4+ T-cell proliferative capacity. The increase in HIV- and herpes-virus-specific CD4+ T-cell proliferative capacity after 55 months of HAART suggests that the improved proliferative response is not specific for HIV, but reflects a more general improvement of antiviral immune responses, which is induced by HAART.


Asunto(s)
Terapia Antirretroviral Altamente Activa , Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , VIH-1/inmunología , Activación de Linfocitos/inmunología , Linfocitos T CD8-positivos/inmunología , Enfermedad Crónica , Citomegalovirus/inmunología , Herpesvirus Humano 4/inmunología , Humanos , Masculino , Factores de Tiempo , Resultado del Tratamiento
19.
PLoS One ; 11(5): e0154342, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27152938

RESUMEN

During hematopoiesis, transcriptional programs are essential for the commitment and differentiation of progenitors into the different blood lineages. GATA1 is a transcription factor expressed in several hematopoietic lineages and essential for proper erythropoiesis and megakaryopoiesis. Megakaryocyte-specific genes, such as GP1BA, are known to be directly regulated by GATA1. Mutations in GATA1 can lead to dyserythropoietic anemia and pseudo gray-platelet syndrome. Selective loss of Gata1 expression in adult mice results in macrothrombocytopenia with platelet dysfunction, characterized by an excess of immature megakaryocytes. To specifically analyze the impact of Gata1 loss in mature committed megakaryocytes, we generated Gata1-Lox|Pf4-Cre mice (Gata1cKOMK). Consistent with previous findings, Gata1cKOMK mice are macrothrombocytopenic with platelet dysfunction. Supporting this notion we demonstrate that Gata1 regulates directly the transcription of Syk, a tyrosine kinase that functions downstream of Clec2 and GPVI receptors in megakaryocytes and platelets. Furthermore, we show that Gata1cKOMK mice display an additional aberrant megakaryocyte differentiation stage. Interestingly, these mice present a misbalance of the multipotent progenitor compartment and the erythroid lineage, which translates into compensatory stress erythropoiesis and splenomegaly. Despite the severe thrombocytopenia, Gata1cKOMK mice display a mild reduction of TPO plasma levels, and Gata1cKOMK megakaryocytes show a mild increase in Pf4 mRNA levels; such a misbalance might be behind the general hematopoietic defects observed, affecting locally normal TPO and Pf4 levels at hematopoietic stem cell niches.


Asunto(s)
Compartimento Celular , Factor de Transcripción GATA1/genética , Megacariocitos/citología , Animales , Ensayo de Inmunoadsorción Enzimática , Megacariocitos/metabolismo , Ratones , Ratones Noqueados , Transcripción Genética
20.
AIDS ; 19(11): 1145-54, 2005 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-15990567

RESUMEN

BACKGROUND: It has been reported that antiretroviral therapy (HAART) during acute HIV-1 infection may rescue HIV-1-specific CD4 T cell responses. OBJECTIVE: To determine the duration of this preserved response by investigating the long-term effects of HAART during acute infection on HIV-specific CD4 T cell function related to possible immune control during subsequent therapy interruption. METHODS: A longitudinal analysis followed HIV-specific CD4 T cell reactivity in 17 individuals with well-documented acute HIV-1 infection where five out of 11 HAART-treated patients stopped therapy and six were untreated. Peripheral blood mononuclear cells were stimulated with overlapping peptide pools derived from Gag and Nef. Production of interferon-gamma (IFN-gamma) and interleukin-2 (IL-2) by CD4 T cells was analysed together with proliferative responses. RESULTS: Absolute numbers, but not percentages, of Gag-specific IFN-gamma-, IL-2- or IFN-gamma/IL-2-producing CD4 T cells were increased in treated compared with untreated individuals up to 2 years after seroconversion. HAART during acute HIV-1 infection was associated with lower viral load but did not result in increased proliferation of HIV-specific CD4 T cells. One out of five individuals who discontinued therapy showed evidence for immune control. However, patients who failed to control viraemia also had measurable proliferative HIV-specific CD4 T cell responses and preserved numbers of cytokine-producing CD4 T cells. CONCLUSIONS: Early HAART during acute HIV-1 infection resulted in higher numbers of HIV-specific IFN-gamma- and IL-2-producing CD4 T cells, but this preservation in four out of five patients was not associated with control of viraemia upon treatment interruption.


Asunto(s)
Terapia Antirretroviral Altamente Activa , Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/tratamiento farmacológico , VIH-1/inmunología , Enfermedad Aguda , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular , Infecciones por VIH/inmunología , Humanos , Inmunidad Celular/inmunología , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Estudios Prospectivos , ARN Viral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA