Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters

Country/Region as subject
Publication year range
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 ; 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
3.
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
4.
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
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.
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
7.
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
8.
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
9.
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
11.
Zhongguo Dang Dai Er Ke Za Zhi ; 10(5): 651-5, 2008 Oct.
Article in Zh | 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
12.
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
13.
Thromb Haemost ; 98(2): 368-74, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17721619

ABSTRACT

The extrinsic coagulation system initiated by tissue factor (TF) appears to be a major procoagulant stimulus during cardiopulmonary bypass (CPB), although the precise mechanisms remain to be revealed. We recently reported the appearance of TF-bearing leukocytes during CPB and described their role in promoting coagulation. In this study, we visually identified the in-vivo appearance of TF-bearing leukocytes and platelet-derived particles on leukocytes in the pericardial blood during cardiac surgery with CPB, by flow cytometry and immunoelectron microscopy. Preliminary flow cytometric experiments showed that the proportion of TF-positive or both TF- and platelet antigen CD41a-positive leukocytes was increased markedly in pericardial blood obtained during CPB, compared with the proportions in preoperative circulating blood. Immunoelectron microscopic analysis revealed that both monocytes and polymorphonuclear leukocytes in the pericardial blood express TF. On the surfaces of these cells, CD41a-positive or both CD41a- and TF-positive platelet-derived particles were observed. Platelet-derived particles include not only microparticles, but also platelets themselves. Leukocytes from preoperative circulating blood contained far fewer of these particles. Our results demonstrate the in-vivo appearance of TF-bearing platelet-derived particles on leukocytes during cardiac surgery with CPB. These findings may be important for the development of strategies to control procoagulant activities during and after cardiac surgery.


Subject(s)
Blood Platelets/metabolism , Cardiopulmonary Bypass , Leukocytes/metabolism , Membrane Microdomains/metabolism , Platelet Membrane Glycoprotein IIb/analysis , Thromboplastin/analysis , Blood Platelets/chemistry , Blood Platelets/ultrastructure , Flow Cytometry , Humans , Leukocytes/chemistry , Microscopy, Immunoelectron , Monocytes/chemistry , Neutrophils/chemistry , Particle Size
14.
Transfus Apher Sci ; 36(3): 285-9, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17602871

ABSTRACT

Platelet concentrates are routinely manufactured from whole blood by differential centrifugation (random donor platelets-RDP) or by plateletpheresis (single donor platelets-SDP). These platelet concentrates have a storage period of 5 days and many different approaches exist to measure the condition of platelets during their storage. In this study, platelet aggregation testing using adenosine diphosphate (ADP) and collagen and flow cytometric platelet activation analysis using CD41 FITC and CD62 PE before and after ADP was performed on days 1, 3 and 5 of storage of platelet preparations. Thirty three RDPs, stored in Baxter and Kansuk blood bags and 18 SDPs stored in Fresenius blood bags were evaluated. In RDPs and in SDPs; ADP and collagen induced PA responses were decreased significantly on the 3rd and 5th days compared to 1st day. CD62 positive platelet percentage after ADP were decreased significantly on the 3rd and 5th days compared to the 1st day in Kansuk bags. Flow cytometric analysis revealed minor changes in CD41 expression after ADP on the 3rd day compared to 1st day and on the 5th day compared to 3rd day. Differences in CD62 positive platelet percentage were not significant between the RDPs and SDPs. Our results suggest that: (1) ADP and collagen induced PA responses decrease both in RDPs and SDPs during storage. (2) Flow cytometric analysis does not show major significant changes in platelet activation after ADP during storage. (3) Continous shaking on the agitator does not cause a significant change in CD62 positive platelet percentage during storage. (4) Platelet aggregation responses in RDPs stored in Baxter and Kansuk blood bags do not differ during storage.


Subject(s)
Platelet Activation , Platelet Function Tests/methods , Plateletpheresis/methods , Adenosine Diphosphate/pharmacology , Blood Platelets/cytology , Blood Preservation/methods , Collagen/pharmacology , Flow Cytometry , Humans , P-Selectin/analysis , Platelet Activation/drug effects , Platelet Membrane Glycoprotein IIb/analysis , Plateletpheresis/standards , Time Factors , Tissue Donors
15.
Thromb Res ; 117(6): 705-11, 2006.
Article in English | MEDLINE | ID: mdl-15990159

ABSTRACT

Platelet-derived microparticles (PMPs) are considered a marker of platelet activation. They vary considerably in size, and flow cytometry, the predominant method used to assay PMPs, is only detecting larger PMPs (>0.1 microm). We describe here a method that quantifies the amount of PMP-located GPIIb antigen in detergent-treated platelet-free plasma (PPP) by means of a one-step time-resolved immunofluorometric assay (TR-IFMA). This assay uses a streptavidin-coated microwell plate and two different monoclonal antibodies to GPIIb (CD41), one conjugated to biotin and the other labeled with europium ion. A wide linear range standard curve with low background and a high sensitivity was obtained. Pre-assay ultracentrifugation or filtration of PPP extensively reduced the fluorometric signal, indicating that the GPIIb antigen is mainly particle-located. A strong correlation between the amount of GPIIb and PMP as detected by flow cytometry was found. Consequently, the assay can be used to study PMP-related phenomena and, in contrast to flow cytometry, can be used on frozen samples and is independent of PMP size.


Subject(s)
Blood Platelets/chemistry , Fluoroimmunoassay , Platelet Activation/physiology , Platelet Membrane Glycoprotein IIb/analysis , Antibodies, Monoclonal/metabolism , Biomarkers/blood , Biotin/metabolism , Biotinylation , Calcimycin/pharmacology , Detergents/pharmacology , Europium/metabolism , Flow Cytometry , Humans , Ionophores/pharmacology , Isotopes , Particle Size , Peptide Fragments/pharmacology , Platelet Activation/drug effects , Sensitivity and Specificity , Streptavidin/metabolism , beta-Thromboglobulin/analysis
16.
Int J Cardiol ; 109(3): 359-66, 2006 May 24.
Article in English | MEDLINE | ID: mdl-16084610

ABSTRACT

BACKGROUND: Oestrogen alters megakaryocyte number in humans and mice. In mice, high-dose oestrogen stimulates an initial increase in megakaryocyte number followed by a decrease. However, the cellular action, effect of physiologically circulating and smaller supra-physiological oestrogen doses and whether changes in megakaryocyte number alter platelet counts have not been studied. METHODS: To further examine oestrogen's effect on megakaryocytes and platelets we administered intact or ovariectomised mice various doses of oestrogen and measured megakaryocyte and platelet counts. To determine the cellular mechanism by which oestrogen influences megakaryocytopoesis we also examined its effect on markers of megakaryocytic differentiation (CD41, CD61, CD34). RESULTS: We found that large doses of oestrogen (500 microg/kg) increased mature CD41+ megakaryocyte number within 2 days, and this was associated with an increase in circulating platelets. Smaller supra-physiological doses (100 microg/kg) lacked this anabolic effect, but still suppressed megakaryocyte and platelet number by day 10 in intact and ovariectomised mice. This was preceded by a reduction in the number of CD61+ megakaryoblasts and CD34+ precursors available to form mature megakaryocytes. In contrast, ovariectomy had no effect on megakaryocyte or platelet number, indicating that circulating oestrogen concentrations do not influence megakaryocyte differentiation or activity. CONCLUSIONS: Our data suggest that in mice at least platelet counts reflect changes in megakaryocyte number, and while both are independent of physiological hormone concentrations, they are sensitive to even small supra-physiological doses of oestrogen. Therefore, to ovoid disrupting platelet homeostasis the dose of oestrogen given should be no more than replacement.


Subject(s)
Estrogens/pharmacology , Megakaryocytes/drug effects , Platelet Count , Animals , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Female , Megakaryocytes/cytology , Mice , Ovariectomy , Platelet Membrane Glycoprotein IIb/analysis
17.
Exp Hematol ; 33(4): 403-12, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15781330

ABSTRACT

OBJECTIVES: GPIIb integrin expression has been found on platelets and megakaryocytes, and more recently on immature hematopoietic progenitors. We set out to investigate expression of GPIIb in other hematopoietic cell lineages and, having detected it on mast cells, aimed to determine what possible role it might perform. METHODS: We have made use of cultured human and murine bone marrow mast cells (BMMC) in order to characterize the expression of GPIIb. Further, BMMC cultures from wild type and GPIIb deficient (gpIIb-/-) mice were used for comparison of the adhesive properties mediated by this receptor. Finally, peritoneal mast cells were analyzed from both wild type and (gpIIb-/-) mice. RESULTS: We demonstrate expression of GPIIb on cultured BMMC. Using cells derived from mice homozygous for a null allele of gbIIb we show that the absence of GPIIb has no effect on mast cells with respect to a number of measures of cell growth and differentiation. However, loss of GPIIb on BMMC results in an increase in surface expression of aV integrin, the alternative partner of GPIIIa. CONCLUSION: The results in this study demonstrate that GPIIb is expressed in human and murine mast cells. A function for GPIIb on mast cells is suggested by the altered adhesion of gbIIb-/- BMMC to fibronectin- and vitronectin-coated surfaces. Moreover, comparison of mast cells from the peritoneal cavity of wild type and gbIIb-/- mice indicates that GPIIb could influence the in vivo differentiation or homing of tissue mast cells.


Subject(s)
Mast Cells/cytology , Platelet Membrane Glycoprotein IIb/genetics , Platelet Membrane Glycoprotein IIb/physiology , Animals , Bone Marrow Cells/cytology , Cell Adhesion , Cell Differentiation , Cell Movement , Cells, Cultured , Fibronectins/metabolism , Humans , Integrin alphaV/analysis , Integrin alphaV/genetics , Mast Cells/chemistry , Mice , Mice, Knockout , Platelet Membrane Glycoprotein IIb/analysis , Vitronectin/metabolism
18.
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
19.
Thromb Haemost ; 94(3): 585-92, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16268476

ABSTRACT

Factor V is an essential coagulation cofactor that circulates in plasma and platelet alpha-granules where it is stored complexed to multimerin I (MMRN1). To gain insights into the origin and processing of human platelet factor V, and factor V-MMRN I complexes, we studied factorV in cultured megakaryocytes. Factor V mRNA was detected in all megakaryocyte cultures. However, like albumin, IgG and fibrinogen, factorV protein was detectable only in megakaryocytes cultured with exogenous protein. The amount of factor V associated with megakaryocytes was influenced by the exogenous factorV concentration. Similar to platelet factor V, megakaryocyte factor V was proteolyzed and complexed with megakaryocyte-synthesized MMRN1. With secretagogues, megakaryocytes released factor V, IgG, fibrinogen and MMRN1. Immunofluorescent and electron microscopy confirmed factorV uptake by endocytosis and its trafficking to megakaryocyte alpha-granules. These data provide direct evidence that human megakaryocytes process plasma-derived factor V into alpha-granules and generate factorV-MMRN I complexes from endogenously and exogenously synthesized proteins.


Subject(s)
Endocytosis , Factor V/metabolism , Megakaryocytes/metabolism , RNA, Messenger/metabolism , Blood Platelets/metabolism , Blood Proteins/metabolism , Bone Marrow Cells/metabolism , Cells, Cultured , Cytoplasmic Granules/metabolism , Factor V/genetics , Fibrinogen/metabolism , Humans , Immunoglobulin G/metabolism , Plasma/metabolism , Platelet Membrane Glycoprotein IIb/analysis , Time Factors
20.
Exp Hematol ; 30(9): 1051-60, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12225797

ABSTRACT

OBJECTIVE: To evaluate the megakaryocyte potential of normal bone marrow (NBM) CD34(+)CD133(+) cells, a subset offering a possible alternative for clinical CD34 immunoselection, we evaluated their colony-forming unit megakaryocyte (CFU-Mk) content and their ability to produce clonogenic Mk progenitors in comparison with the CD133(-) subset. MATERIALS AND METHODS: Sorted NBM CD34(+)CD133(+) and CD34(+)CD133(-) subsets were evaluated for Mk clonogenic capacity before and after in vitro proliferation in serum-free liquid culture containing kit ligand, Flt3 ligand, thrombopoietin, interleukin-3, and interleukin-6. The segregation of CFU-Mk according to the expression of CD34, CD133, and CD41 was compared between fresh BM cells and expanded cells. RESULTS: Although the fresh NBM CD133(-)CD34(+) subset included two thirds CFU-Mk, only the CD133(+) subset contained primitive cells able to produce all categories of CFU-Mk in vitro. Immunophenotyping confirmed that CD41 antigen is nonspecific for Mk lineage and showed that the usual CD34(+)CD41(+) subset does not specifically define a CFU-Mk population. The segregation of CFU-Mk before and after expansion according to CD34, CD41, or CD133 was modified in relation with down-regulation of CD34 and CD133 antigens and up-regulation of CD41 antigen. CONCLUSIONS: The NBM CD133(+) subset contains primitive cells able to generate CFU-Mk, a subset probably relevant to platelet recovery after infusion. The alteration of antigen expression during in vitro proliferation calls for caution in the identification of the different categories of Mk subsets produced and in the assessment of their predictivity for in vivo platelet production.


Subject(s)
Bone Marrow Cells/cytology , Hematopoietic Stem Cells/cytology , Megakaryocytes/cytology , AC133 Antigen , Antigens, CD , Antigens, CD34/analysis , Cell Differentiation , Cell Lineage , Cell Separation , Cells, Cultured/cytology , Colony-Forming Units Assay , Culture Media, Serum-Free , Erythroid Precursor Cells/cytology , Glycoproteins/analysis , Granulocytes/cytology , Humans , Megakaryocytes/classification , Peptides/analysis , Platelet Membrane Glycoprotein IIb/analysis
SELECTION OF CITATIONS
SEARCH DETAIL