Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Blood ; 138(14): 1211-1224, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34115843

ABSTRACT

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.


Subject(s)
Mice/genetics , Single-Cell Analysis , Thrombopoiesis , Transcriptome , Animals , Cells, Cultured , Female , Humans , Male , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice/physiology , Mice, Inbred C57BL , Platelet Membrane Glycoprotein IIb/analysis , Platelet Membrane Glycoprotein IIb/genetics , Ploidies
2.
Blood Coagul Fibrinolysis ; 31(5): 310-316, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32398462

ABSTRACT

: Autophagy is a conserved cellular process that involves the degradation of cytoplasmic components in eukaryotic cells. However, the correlation between autophagy and megakaryocyte development is unclear. This study aims to explore the role of autophagy in megakaryocyte differentiation. To test our hypothesis, we used the Dami cell line in-vitro experiments. Rapamycin and Bafilomycin A1 were used to stimulate Dami cells. CD41 expression and apoptosis were analysed by flow cytometry. Autophagy-related proteins were detected by Western blotting. 12-O-Tetradecanoylphorbol 13-acetate-treated Dami cells can simulate endomitosis of megakaryocytes in vitro. Rapamycin-induced autophagic cell death was verified by LC3-II conversion upregulation. Meanwhile, Bafilomycin A1 blocked endomitosis and autophagy of Dami cells. Our results provide evidence that autophagy is involved in megakaryocyte endomitosis and platelet development. Rapamycin inhibited cell viability and induced multiple cellular events, including apoptosis, autophagic cell death, and megakaryocytic differentiation, in human Dami cells. Upregulated autophagy triggered by rapamycin can promote the differentiation of Dami cells, while endomitosis is accompanied by enhanced autophagy.


Subject(s)
Autophagy/drug effects , Cell Differentiation/drug effects , Megakaryocytes/drug effects , Sirolimus/pharmacology , Apoptosis/drug effects , Cell Line , Humans , Macrolides/pharmacology , Megakaryocytes/cytology , Platelet Membrane Glycoprotein IIb/analysis
3.
An. Facultad Med. (Univ. Repúb. Urug., En línea) ; 6(2): 25-34, dic. 2019. ilus, graf
Article in Spanish | LILACS, UY-BNMED, BNUY | ID: biblio-1291263

ABSTRACT

El estudio de la megacariopoyesis humana se ha visto obstaculizado por la relativa escasez de megacariocitos en la médula ósea (0,05-0,2 % de las células medulares), lo que ha llevado a la optimización de protocolos de expansión in vitro a partir de precursores de diversos orígenes (cordón umbilical, médula ósea y sangre periférica con o sin movilización previa). Los cultivos celulares a partir de precursores han permitido la producción y el estudio tanto de megacariocitos así como de proplaquetas y plaquetas Sin embargo, la producción in vitro óptima de megacariocitos que culminen todos los estadios de diferenciación es un reto aún no resuelto. En este trabajo reportamos los hallazgos concernientes a la determinación de las condiciones y concentraciones de trombopoyetina para lograr una óptima relación entre la cantidad de trombopoyetina empleada y el porcentaje y grado de diferenciación megacariocítica en muestras obtenidas de cinco donantes alogénicos aceptados para trasplante de médula ósea.


The study of human megakaryocytopoiesis has been hampered by the relative scarcity of megakaryocytes in bone marrow (0.05-0.2 % of medullary cells), which has led to the optimization of protocols of in vitro expansion of precursors from diverse sources (umbilical cord, bone marrow and peripheral blood with or without previous mobilization). Cell cultures from different precursors have allowed the production and study of megakaryocytes as well as proplatelets and platelets. However, the in vitro production of megakaryocytes that culminate all stages of differentiation is a challenge that has not yet been resolved. In this work we report the findings related to the determination of thrombopoietin treatment conditions and concentrations to achieve an optimal relationship between the amount of thrombopoietin and the percentage and degree of megakaryocytic differentiation in five allogeneic donors that were accepted for bone marrow transplantation.


O estudo da megacariopoiese humana tem sido dificultado pela relativa escassez de megacariócitos na medula óssea (0,05-0,2 % das células medulares), o que levou à otimização dos protocolos de expansão in vitro a partir de precursores de diversas origens (cordão umbilical, medula óssea e sangue periférico com ou sem mobilização prévia). Culturas de células a partir de precursores permitiram a produção e o estudo tanto de megacariócitos e de proplaquetas e plaquetas. No entanto, a produção ótima in vitro de megacariócitos que culminam em todas as fases de diferenciação é um desafio ainda não resolvido. Neste trabalho, relatamos as descobertas relativas à determinação das condições e concentrações de trombopoietina para obter uma relação ótima entre a quantidade de trombopoietina usada e a taxa e o grau de diferenciação megacariocítica em amostras obtidas de cinco doadores alogênicos aceitos para transplante de medula óssea.


Subject(s)
Humans , Thrombopoietin/analysis , Megakaryocytes/cytology , Antigens, CD34/analysis , Cells, Cultured/cytology , Leukapheresis , Platelet Membrane Glycoprotein IIb/analysis , Integrin beta3/analysis , Culture Techniques/methods
4.
Blood ; 129(25): 3332-3343, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28336526

ABSTRACT

The developmental pathway for human megakaryocytes remains unclear, and the definition of pure unipotent megakaryocyte progenitor is still controversial. Using single-cell transcriptome analysis, we have identified a cluster of cells within immature hematopoietic stem- and progenitor-cell populations that specifically expresses genes related to the megakaryocyte lineage. We used CD41 as a positive marker to identify these cells within the CD34+CD38+IL-3RαdimCD45RA- common myeloid progenitor (CMP) population. These cells lacked erythroid and granulocyte-macrophage potential but exhibited robust differentiation into the megakaryocyte lineage at a high frequency, both in vivo and in vitro. The efficiency and expansion potential of these cells exceeded those of conventional bipotent megakaryocyte/erythrocyte progenitors. Accordingly, the CD41+ CMP was defined as a unipotent megakaryocyte progenitor (MegP) that is likely to represent the major pathway for human megakaryopoiesis, independent of canonical megakaryocyte-erythroid lineage bifurcation. In the bone marrow of patients with essential thrombocythemia, the MegP population was significantly expanded in the context of a high burden of Janus kinase 2 mutations. Thus, the prospectively isolatable and functionally homogeneous human MegP will be useful for the elucidation of the mechanisms underlying normal and malignant human hematopoiesis.


Subject(s)
Hematopoiesis , Megakaryocyte Progenitor Cells/cytology , Megakaryocyte Progenitor Cells/metabolism , Megakaryocytes/cytology , Adult , Animals , Antigens, CD/analysis , Cell Lineage , Cells, Cultured , Humans , Megakaryocyte Progenitor Cells/pathology , Megakaryocytes/metabolism , Mice, Inbred C57BL , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Platelet Membrane Glycoprotein IIb/analysis , Transcriptome
5.
Transfusion ; 56(9): 2286-95, 2016 09.
Article in English | MEDLINE | ID: mdl-27443848

ABSTRACT

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.


Subject(s)
Blood Platelets/metabolism , RNA, Messenger/genetics , Riboflavin/pharmacology , Ultraviolet Rays , Blood Platelets/drug effects , Blood Platelets/radiation effects , Blood Preservation/methods , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/analysis , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , Humans , Integrin beta3/analysis , Integrin beta3/genetics , Osteonectin/analysis , Osteonectin/genetics , Platelet Factor 4/analysis , Platelet Factor 4/genetics , Platelet Membrane Glycoprotein IIb/analysis , Platelet Membrane Glycoprotein IIb/genetics , RNA, Messenger/drug effects , RNA, Messenger/radiation effects , Thrombospondins/analysis , Thrombospondins/genetics
6.
J Hematol Oncol ; 9(1): 45, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27177927

ABSTRACT

BACKGROUND: Somatic calreticulin (CALR), Janus kinase 2 (JAK2), and thrombopoietin receptor (MPL) mutations essentially show mutual exclusion in myeloproliferative neoplasms (MPN), suggesting that they activate common oncogenic pathways. Recent data have shown that MPL function is essential for CALR mutant-driven MPN. However, the exact role and the mechanisms of action of CALR mutants have not been fully elucidated. METHODS: The murine myeloid cell line 32D and human HL60 cells overexpressing the most frequent CALR type 1 and type 2 frameshift mutants were generated to analyze the first steps of cellular transformation, in the presence and absence of MPL expression. Furthermore, mutant CALR protein stability and secretion were examined using brefeldin A, MG132, spautin-1, and tunicamycin treatment. RESULTS: The present study demonstrates that the expression of endogenous Mpl, CD41, and the key megakaryocytic transcription factor NF-E2 is stimulated by type 1 and type 2 CALR mutants, even in the absence of exogenous MPL. Mutant CALR expressing 32D cells spontaneously acquired cytokine independence, and this was associated with increased Mpl mRNA expression, CD41, and NF-E2 protein as well as constitutive activation of downstream signaling and response to JAK inhibitor treatment. Exogenous expression of MPL led to constitutive activation of STAT3 and 5, ERK1/2, and AKT, cytokine-independent growth, and reduction of apoptosis similar to the effects seen in the spontaneously outgrown cells. We observed low CALR-mutant protein amounts in cellular lysates of stably transduced cells, and this was due to accelerated protein degradation that occurred independently from the ubiquitin-proteasome system as well as autophagy. CALR-mutant degradation was attenuated by MPL expression. Interestingly, we found high levels of mutated CALR and loss of downstream signaling after blockage of the secretory pathway and protein glycosylation. CONCLUSIONS: These findings demonstrate the potency of CALR mutants to drive expression of megakaryocytic differentiation markers such as NF-E2 and CD41 as well as Mpl. Furthermore, CALR mutants undergo accelerated protein degradation that involves the secretory pathway and/or protein glycosylation.


Subject(s)
Calreticulin/genetics , Cell Transformation, Neoplastic/genetics , Golgi Apparatus/metabolism , Megakaryocytes/metabolism , Mutant Proteins/physiology , Signal Transduction , Animals , Calreticulin/physiology , Cell Death , Cell Line , Cell Line, Tumor , Frameshift Mutation , Humans , Mice , Myeloproliferative Disorders/genetics , NF-E2 Transcription Factor, p45 Subunit/analysis , Platelet Membrane Glycoprotein IIb/analysis , Proteolysis , Receptors, Thrombopoietin/analysis
7.
Blood ; 127(18): 2231-40, 2016 05 05.
Article in English | MEDLINE | ID: mdl-26966088

ABSTRACT

The mechanisms regulating megakaryopoiesis and platelet production (thrombopoiesis) are still incompletely understood. Identification of a progenitor with enhanced thrombopoietic capacity would be useful to decipher these mechanisms and to improve our capacity to produce platelets in vitro. Differentiation of peripheral blood CD34(+) cells in the presence of bone marrow-human mesenchymal stromal cells (MSCs) enhanced the production of proplatelet-bearing megakaryocytes (MKs) and platelet-like elements. This was accompanied by enrichment in a MK precursor population exhibiting an intermediate level of CD41 positivity while maintaining its expression of CD34. Following sorting and subculture with MSCs, this CD34(+)CD41(low) population was able to efficiently generate proplatelet-bearing MKs and platelet-like particles. Similarly, StemRegenin 1 (SR1), an antagonist of the aryl hydrocarbon receptor (AhR) transcription factor known to maintain CD34 expression of progenitor cells, led to an enriched CD34(+)CD41(low) fraction and to an increased capacity to generate proplatelet-producing MKs and platelet-like elements ultrastructurally and functionally similar to circulating platelets. The effect of MSCs, like that of SR1, appeared to be mediated by an AhR-dependent mechanism because both culture conditions resulted in repression of its downstream effector CYP1B1. This newly described isolation of a precursor exhibiting strong MK potential could be exploited to study normal and abnormal thrombopoiesis and for in vitro platelet production.


Subject(s)
Megakaryocyte Progenitor Cells/cytology , Receptors, Aryl Hydrocarbon/physiology , Thrombopoiesis/physiology , Antigens, CD34/analysis , Blood Platelets/cytology , Cell Separation , Cells, Cultured , Coculture Techniques , Culture Media, Serum-Free , Cytochrome P-450 CYP1B1/physiology , Humans , Immunophenotyping , Platelet Count , Platelet Membrane Glycoprotein IIb/analysis , Purines/pharmacology , Signal Transduction
8.
Theranostics ; 5(4): 418-30, 2015.
Article in English | MEDLINE | ID: mdl-25699100

ABSTRACT

OBJECTIVE: Ultrasound molecular imaging (UMI) of glycoprotein (GP) IIb/IIIa receptor on activated platelets offers a unique means of identifying high-risk atherosclerosis. We hypothesized that contrast-enhanced ultrasound with microbubbles (MBs) targeted to GP IIb/IIIa could be used to detect and quantify activated platelets on the surface of advanced plaques. METHODS AND RESULTS: A mouse model of advanced atherosclerosis was generated by maintaining apolipoprotein E-deficient (ApoE(-/-)) mice on a hypercholesterolemic diet (HCD). The three other experimental groups consisted of ApoE(-/-) and wild-type (C57BL/6) mice fed a normal chow diet and C57BL/6 mice on an HCD diet. Plaque formation was confirmed by histological and immunohistochemical methods using light, fluorescence, and electron microscopy. Mice were injected with a lipid MB-conjugated cyclic Arg-Gly-Asp peptide or nonspecific control peptide, and the abdominal aorta was examined by UMI. The accumulation of GP IIb/IIIa and activated platelets on the surface of atherosclerotic plaques was highest in the ApoE(-/-)+HCD group, followed by ApoE(-/-)+chow, C57BL/6+HCD, and C57BL/6+chow groups (P<0.05). Notably, GP IIb/IIIa expression was associated with the vulnerability index and necrotic center/fiber cap ratio (P<0.05), and contrast video intensity from adhered cyclic Arg-Gly-Asp-modified MBs (MB-cRGDs) was correlated with GP IIb/IIIa expression on the plaque surface (P<0.05). CONCLUSION: GP IIb/IIIa of activated platelets on the atherosclerotic endothelium is a biomarker for high-risk plaques that can be quantified by UMI using MB-cRGDs, providing a noninvasive means for detecting high-risk plaques and preventing acute cardiovascular events.


Subject(s)
Blood Platelets/chemistry , Integrin beta3/analysis , Microbubbles , Molecular Imaging/methods , Plaque, Atherosclerotic/pathology , Platelet Membrane Glycoprotein IIb/analysis , Ultrasonography/methods , Animals , Biomarkers/analysis , Disease Models, Animal , Mice, Inbred C57BL
9.
Blood ; 125(6): 1025-33, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25343958

ABSTRACT

Several approaches for controlling hematopoietic stem and progenitor cell expansion, lineage commitment, and maturation have been investigated for improving clinical interventions. We report here that amino acid substitutions in a thrombopoietin receptor (Mpl)--containing cell growth switch (CGS) extending receptor stability improve the expansion capacity of human cord blood CD34(+) cells in the absence of exogenous cytokines. Activation of this CGS with a chemical inducer of dimerization (CID) expands total cells 99-fold, erythrocytes 70-fold, megakaryocytes 0.5-fold, and CD34(+) stem/progenitor cells 4.4-fold by 21 days of culture. Analysis of cells in these expanded populations identified a CID-dependent bipotent erythrocyte-megakaryocyte precursor (PEM) population, and a CID-independent macrophage population. The CD235a(+)/CD41a(+) PEM population constitutes up to 13% of the expansion cultures, can differentiate into erythrocytes or megakaryocytes, exhibits very little expansion capacity, and exists at very low levels in unexpanded cord blood. The CD206(+) macrophage population constitutes up to 15% of the expansion cultures, exhibits high-expansion capacity, and is physically associated with differentiating erythroblasts. Taken together, these studies describe a fundamental enhancement of the CGS expansion platform, identify a novel precursor population in the erythroid/megakaryocytic differentiation pathway of humans, and implicate an erythropoietin-independent, macrophage-associated pathway supporting terminal erythropoiesis in this expansion system.


Subject(s)
Amino Acid Substitution , Erythroid Cells/cytology , Erythropoiesis , Megakaryocytes/cytology , Receptors, Thrombopoietin/genetics , Animals , Antigens, CD34/analysis , Cell Line , Cell Proliferation , Cells, Cultured , Erythroid Cells/metabolism , Fetal Blood/cytology , Humans , Megakaryocytes/metabolism , Mice , Platelet Membrane Glycoprotein IIb/analysis , Receptors, Thrombopoietin/metabolism
11.
Stem Cells Dev ; 21(14): 2592-605, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22489670

ABSTRACT

CD41 (αIIb integrin), a specific marker for megakaryocytes and platelets, was recently shown to mark the initiation of definitive hematopoiesis in mouse embryos. However, whether embryonic CD41(+) populations have a nonhematopoietic potential remains elusive. Here, we report that the CD41(+) cells from the mouse E11.0 aorta-gonad-mesonephros (AGM) region and yolk sac (YS) expressed a set of mesenchymal markers (as revealed by reverse transcriptase-polymerase chain reaction), displayed myofibroblastic/fibroblastic potential in vitro under mesenchymal culture conditions, and differentiated into α-SMA(+)/epimorphin(+)/vimentin(+) cells in the lungs of adult recipients after systemic transplantation. This unique cell population with fibroblastic potential expressed intermediate rather than high levels of CD41 and was negative for CD34 in the AGM region. In contrast, circulating CD41(+) cells in the embryonic blood stream harbored no similar fibroblastic potential. Compared with the YS, the AGM-derived CD41(+) cells had a more robust fibroblastic potential, as revealed by higher in vitro growth rates. Interestingly, the AGM-derived CD41(+) cells demonstrated a stronger response to the chemotaxin of circulating blood plasma than the YS-derived CD41(+) cells. We are the first group that illustrates the fibroblastic potential of an embryonic CD41(+) population in vitro and in vivo, reflecting the close association between blood and mesenchyme development during mouse mid-gestation. The precise origin of these mesenchymal populations needs further clarification.


Subject(s)
Aorta/cytology , Fibroblasts/cytology , Gonads/cytology , Mesonephros/cytology , Platelet Membrane Glycoprotein IIb/analysis , Yolk Sac/cytology , Actins/metabolism , Animals , Antigens, CD34 , Aorta/embryology , Biomarkers/analysis , Cell Culture Techniques , Cell Differentiation , Cell Migration Assays , Cell Transplantation/methods , Colony-Forming Units Assay , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Female , Fibroblasts/transplantation , Gonads/embryology , Green Fluorescent Proteins , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Lung/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Vimentin/metabolism , Whole-Body Irradiation , Yolk Sac/embryology
12.
Methods Mol Biol ; 750: 143-55, 2011.
Article in English | MEDLINE | ID: mdl-21618089

ABSTRACT

The use of transgenic mice in which tissue or lineage-specific, cell-restricted promoters drive fluorescent reporters has recently been reported as a means to follow the in vivo migration of various hematopoietic cells during murine development. At present there is limited ability of these approaches to image the emergence of the first hematopoietic cell subsets due to lack of unique markers that define those hematopoietic cells. We have utilized whole embryo analysis via immunostaining and confocal laser-scanning microscopic (CLSM) imaging to define the emergence of the first hematopoietic elements in the yolk sac of the developing conceptus. The methods employed to examine yolk sac hematopoiesis may be applied to hematopoietic cell emergence in the embryo proper or fetal liver in the generation of a complete map of hematopoietic ontogeny.


Subject(s)
Cell Movement , Embryo, Mammalian/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Platelet Membrane Glycoprotein IIb/analysis , Whole Body Imaging/methods , Yolk Sac/cytology , Animals , Cell Lineage , Embryo, Mammalian/cytology , Fluorescence , Genes, Reporter , Green Fluorescent Proteins/analysis , Immunochemistry , Mice , Mice, Transgenic , Microscopy, Confocal , Platelet Membrane Glycoprotein IIb/biosynthesis , Staining and Labeling/methods , Yolk Sac/embryology , Yolk Sac/physiology
14.
Thromb Res ; 127(4): 370-7, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21257195

ABSTRACT

INTRODUCTION: Circulating cell-derived microparticles (MPs) have been implicated in several disease processes and elevated levels are found in many pathological conditions. The detection and accurate measurement of MPs, although attracting widespread interest, is hampered by a lack of standardisation. The aim of this study was to establish a reliable flow cytometric assay to measure distinct subtypes of MPs in disease and to identify any significant causes of variability in MP quantification. MATERIALS AND METHODS: Circulating MPs within plasma were identified by their phenotype (platelet, endothelial, leukocyte and annexin-V positivity (AnnV+). The influence of key variables (i.e. time between venepuncture and centrifugation, washing steps, the number of centrifugation steps, freezing/long-term storage and temperature of thawing) on MP measurement were investigated. RESULTS: Increasing time between venepuncture and centrifugation leads to increased MP levels. Washing samples results in decreased AnnV+MPs (P=0.002) and platelet-derived MPs (PMPs) (P=0.002). Double centrifugation of MPs prior to freezing decreases numbers of AnnV+MPs (P=0.0004) and PMPs (P=0.0004). A single freeze thaw cycle of samples led to an increase in AnnV+MPs (P=0.0020) and PMPs (P=0.0039). Long-term storage of MP samples at -80° resulted in decreased MP levels. CONCLUSIONS: This study found that minor protocol changes significantly affected MP levels. This is one of the first studies attempting to standardise a method for obtaining and measuring circulating MPs. Standardisation will be essential for successful development of MP technologies, allowing direct comparison of results between studies and leading to a greater understanding of MPs in disease.


Subject(s)
Cell-Derived Microparticles/metabolism , Flow Cytometry/methods , Aged , Annexin A5/analysis , Blood Platelets/cytology , Endothelial Cells/cytology , Humans , Leukocytes/cytology , Middle Aged , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Platelet Membrane Glycoprotein IIb/analysis , Reproducibility of Results
15.
Biochem Biophys Res Commun ; 405(1): 128-33, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21216234

ABSTRACT

Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) is an inhibitory collagen receptor which belongs to the immunoglobulin (Ig) superfamily. Although the inhibitory function of LAIR-1 has been extensively described in multiple leukocytes, its role in megakaryocyte (MK) has not been explored so far. Here, we show that LAIR-1 is expressed on human bone marrow CD34(+)CD41a(+) and CD41a(+)CD42b(+) cells. LAIR-1 is also detectable in a fraction of human cord blood CD34(+) cell-derived MK that has morphological characteristics of immature MK. In megakaryoblastic cell line Dami, the membrane protein expression of LAIR-1 is up-regulated significantly when cells are treated with phorbol ester phorbol 12-myristate 13-acetate (PMA). Furthermore, cross-linking of LAIR-1 in Dami cells with its natural ligand or anti-LAIR-1 antibody leads to the inhibition of cell proliferation and PMA-promoted differentiation when examined by the MK lineage-specific markers (CD41a and CD42b) and polyploidization. In addition, we also observed that cross-linking of LAIR-1 results in decreased MK generation from primary human CD34(+) cells cultured in a cytokines cocktail that contains TPO. These results suggest that LAIR-1 is a likely candidate for an early marker of MK differentiation, and provide initial evidence indicating that LAIR-1 serves as a negative regulator of megakaryocytopoiesis.


Subject(s)
Cell Differentiation , Megakaryocytes/cytology , Receptors, Immunologic/metabolism , Stem Cells/cytology , Antigens, CD34/analysis , Antigens, CD34/metabolism , Biomarkers , Cell Line , Fetal Blood/cytology , Humans , Megakaryocytes/metabolism , Platelet Membrane Glycoprotein IIb/analysis , Platelet Membrane Glycoprotein IIb/metabolism , Receptors, Immunologic/analysis , Stem Cells/chemistry , Stem Cells/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Up-Regulation
16.
Rheumatology (Oxford) ; 48(8): 1003-7, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19542214

ABSTRACT

OBJECTIVES: To evaluate whether the use of platelet immunohistochemistry (IHC) markers improves the sensitivity of histological methods to detect microthrombosis in SLE nephritis and aPLs and to analyse the clinicopathological correlations of microthrombosis in this setting. METHODS: Kidney biopsy specimens from 65 patients with SLE, including 36 with positive aPLs, were studied by IHC using antibodies against platelet glycoproteins CD41 and CD61. Clinical data at the time of kidney biopsy and during a mean follow-up of 7.5 years after biopsy were recorded and analysed with regard to histological or IHC data. RESULTS: Histological lesions previously defined as APS nephropathy were found in 33% of the SLE kidney biopsies and were not associated with positive aPLs. Microthrombi detected as intravascular CD61(+) platelet deposits were present in 43% of the tissues and were significantly associated with positive aPLs, but not with histological APS nephropathy, nephritis manifestations nor with renal outcome. Histological APS lesions but not CD61(+) microthrombi correlated with an older age at nephritis presentation, previous cardiovascular risk factors and worse renal outcome. CONCLUSIONS: Immunodetection of intravascular CD61(+) platelet aggregates is more sensitive than histological evaluation to detect acute microthrombosis and provides a better correlation with aPLs in SLE patients. In contrast, histological lesions consistent with APS nephropathy were not associated with aPLs but with cardiovascular risk factors and worse renal outcome.


Subject(s)
Antibodies, Antiphospholipid , Lupus Nephritis/complications , Thrombosis/complications , Adult , Aged , Aged, 80 and over , Biomarkers/analysis , Female , Humans , Immunohistochemistry , Integrin beta3/analysis , Kidney/immunology , Kidney/pathology , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Male , Microvessels , Middle Aged , Odds Ratio , Platelet Aggregation , Platelet Membrane Glycoprotein IIb/analysis , Sensitivity and Specificity , Thrombosis/immunology , Thrombosis/pathology , Young Adult
17.
Vet Immunol Immunopathol ; 131(1-2): 25-32, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19362377

ABSTRACT

Recurrent airway obstruction (RAO) in mature horses is characterized by reversible airway obstruction and neutrophilic inflammation; there is also functional activation of circulating platelets and neutrophils. This study was undertaken to determine if changes in activation marker expression and heterotypic aggregate formation can be used as an indicator of this increased functional responsiveness. In vitro conditions for flow cytometric measurement of CD13, CD41/61 and CD62P expression on activated cells and heterotypic aggregate formation were established. Values were then compared before and after antigen challenge of RAO and healthy horses. Platelet adhesion to serum-coated plastic was measured as a functional marker of platelet activation. In vitro activation resulted in increased expression of neutrophil CD13 and platelet CD41/61 and CD62P. Activation of both cell types caused a significant increase in neutrophil-platelet aggregates. In horses with RAO, but not controls, there was a significant increase in the percentage of CD13 positive neutrophils at 10h and 24h and in the mean fluorescence intensity at 10h. This was accompanied at 24h by an increased mean platelet side scatter and thrombin-stimulated platelet adhesion. In conclusion, CD13 expression can be used as an indicator of equine neutrophil activation both in vitro and in vivo. Equine platelet activation in vitro can be detected by measuring CD41/61 or CD62P expression, and PAF-activated platelets and neutrophils form aggregates. However, despite evidence of circulating platelet activation, neither a change in expression of platelet activation markers, nor heterotypic aggregate aggregate formation could be detected.


Subject(s)
Airway Obstruction/veterinary , CD13 Antigens/analysis , Horse Diseases/immunology , Integrin beta3/analysis , Neutrophils/physiology , P-Selectin/analysis , Platelet Activation , Platelet Aggregation , Platelet Membrane Glycoprotein IIb/analysis , Airway Obstruction/blood , Airway Obstruction/immunology , Animals , Cell Aggregation , Horses , Platelet Adhesiveness , Recurrence
18.
Zhongguo Dang Dai Er Ke Za Zhi ; 10(5): 651-5, 2008 Oct.
Article in Chinese | MEDLINE | ID: mdl-18947492

ABSTRACT

OBJECTIVE: The mechanism of neonatal hypoxic-ischemic brain damage (HIBD) remains unclear and effective treatment approach is limited for this disorder. Many studies have shown that tissue-type plasminogen activator (tPA) plays an important role in nervous system. This study investigated the effect of tPA in HIBD in neonatal rats. METHODS: Seven-day-old Wistar rat pups were used for the Rice-Vannucci model of neonatal hypoxia-ischemia (HI). Brain samples were collected 1, 4, and 24 hrs after HI. FITC-Dextran was injected into the left ventricle of pups after HI to observe reperfusion defects of the neonatal brain. RT-PCR and tPA zymogram were used to detect the expression and activity of tPA. Double immunostaining, terminal deoxynucleotidyl transferase mediated dUTP nick end labeling (TUNEL) and DAPI staining were used to detect the expression of integrin GPIIb and fibrin and neuronal apoptosis. RESULTS: FITC-Dextran perfusion analysis indicated there was obvious infarct area in the neonatal brain and the expression of integrin GPIIb and fibrin increased significantly 1 hr after HI compared with the contralateral side. The infarct area decreased and the expression of integrin GPIIb and fibrin were reduced 4 hrs after HI. The expression and activity of tPA increased significantly in neonatal rats after HI, and peaked at 4 hrs after HI. The number of apoptotic neural cells increased progressively with the prolonged reperfusion time following HI. CONCLUSIONS: The increase of tPA in the acute phase after HIBD may be helpful to clot dissolving, but it induces neuronal apoptosis and aggravates brain injury.


Subject(s)
Hypoxia-Ischemia, Brain/metabolism , Tissue Plasminogen Activator/physiology , Animals , Animals, Newborn , Apoptosis , Fibrin/analysis , Hypoxia-Ischemia, Brain/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Platelet Membrane Glycoprotein IIb/analysis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Tissue Plasminogen Activator/analysis
19.
J Antimicrob Chemother ; 62(5): 1118-21, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18753189

ABSTRACT

INTRODUCTION: Thromboembolic complications under antiretroviral therapy (ART) have been described in the past. In particular, the influence of protease inhibitors (PIs) on platelet activation and coagulation is currently under discussion. METHODS: HIV-1-infected, PI-naive adults (n = 18) were investigated before and 4 weeks after the start of the ART, consisting either of boosted PI regimens (n = 13) plus reverse transcriptase inhibitors (RTIs) or a double PI regimen (n = 5) without RTI co-medication. Administered PIs were saquinavir (n = 15), lopinavir (n = 4), fosamprenavir (n = 2) and atazanavir (n = 2). Platelet CD62P, CD40L (%+ cells) and PAC-1 binding [mean fluorescence intensity (MFI)] as well as monocyte CD11b (MFI) and monocyte-associated CD41 (%+ cells and MFI) expression were assessed by flow cytometry with or without platelet stimulation. To investigate the influence of platelets on coagulation, the endogenous thrombin potential (ETP) [render fluorescence units (RFI)] was determined. RESULTS: CD62P, PAC-1 binding and CD11b expression remained unchanged. In contrast, the mean+/-SD MFI of CD40L (from 18.2+/-9.0 to 25.5+/-10.4, P = 0.038) and CD41 (from 446.1+/-213.8 to 605.0+/-183.8, P = 0.010) as markers for increased platelet-leucocyte interaction increased significantly. The collagen-induced ETP time-to-peak was altered significantly from 23.8+/-11.4 to 17.0+/-4.2 min (P = 0.028), although the ETP RFI peak showed no evidence for increased procoagulatory capacity (47.1+/-18.6 to 57.3+/-19.9, P = 0.085). CONCLUSIONS: Effects of the evaluated PI HIV therapy on platelet function assessed under field conditions seem to be minor, not affecting all investigated parameters. We found no evidence for increased platelet activation under PI-containing ART. However, CD41 as a marker for increased platelet-leucocyte interaction and CD40L, which can contribute to atherosclerosis, increased significantly.


Subject(s)
Blood Platelets/drug effects , Cell Adhesion Molecules/biosynthesis , HIV Infections/drug therapy , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/therapeutic use , Leukocytes/drug effects , Adult , Blood Platelets/chemistry , CD11b Antigen/analysis , CD40 Ligand/analysis , Female , Flow Cytometry , Humans , Leukocytes/chemistry , Male , Middle Aged , P-Selectin/analysis , Platelet Membrane Glycoprotein IIb/analysis
20.
Int J Radiat Oncol Biol Phys ; 69(4): 1272-81, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17967317

ABSTRACT

PURPOSE: Specific genes expressed as a result of whole body exposure to gamma-radiation have been previously identified. In this study, we examined the genes further as possible biomarkers for the blood lymphocytes of C57BL/6 mice after whole body or local irradiation of the thorax, abdomen, and left subphrenic area. METHODS AND MATERIALS: We performed reverse transcriptase-polymerase chain reaction and real-time reverse transcriptase-polymerase chain reaction analysis of genes encoding platelet membrane glycoprotein IIb, protein tyrosine kinase, sialyltransferase, and Cu/ZnSOD in blood lymphocytes, lung tissue, spleen, and intestines. The protein expression in blood lymphocytes was confirmed by Western blot analysis. RESULTS: The expression of platelet membrane glycoprotein IIb, protein tyrosine kinase, sialyltransferase, and Cu/ZnSOD was significantly greater after 3 days as a result of 1 Gy of whole body irradiation. Moreover, local irradiation to the thorax, abdomen, or left subphrenic area, which are frequently exposed to therapeutic radiation doses, showed a tendency toward radiation-induced increased expression of these genes in both the blood and the locally irradiated organs. Western blot analysis also corroborated these results. CONCLUSION: Platelet membrane glycoprotein IIb, protein tyrosine kinase, sialyltransferase, and Cu/ZnSOD might be candidates for biomarkers of radiation exposure. However, additional experiments are required to reveal the relationship between the expression levels and the prognostic effects after irradiation.


Subject(s)
Biomarkers/analysis , Gamma Rays , Gene Expression Profiling/methods , Lymphocytes/radiation effects , Whole-Body Irradiation , Animals , Biomarkers/metabolism , Female , Intestinal Mucosa/metabolism , Intestines/radiation effects , Lung/metabolism , Lung/radiation effects , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Platelet Membrane Glycoprotein IIb/analysis , Platelet Membrane Glycoprotein IIb/metabolism , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sialyltransferases/analysis , Sialyltransferases/genetics , Sialyltransferases/metabolism , Spleen/metabolism , Spleen/radiation effects , Superoxide Dismutase/analysis , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...