scholarly journals Effect of MSCs and MSC-Derived Extracellular Vesicles on Human Blood Coagulation

Cells ◽  
2019 ◽  
Vol 8 (3) ◽  
pp. 258 ◽  
Author(s):  
Denis Silachev ◽  
Kirill Goryunov ◽  
Margarita Shpilyuk ◽  
Olga Beznoschenko ◽  
Natalya Morozova ◽  
...  

Mesenchymal stem cells (MSCs) have emerged as a potent therapeutic tool for the treatment of a number of pathologies, including immune pathologies. However, unwelcome effects of MSCs on blood coagulation have been reported, motivating us to explore the thrombotic properties of human MSCs from the umbilical cord. We revealed strong procoagulant effects of MSCs on human blood and platelet-free plasma using rotational thromboelastometry and thrombodynamic tests. A similar potentiation of clotting was demonstrated for MSC-derived extracellular vesicles (EVs). To offer approaches to avoid unwanted effects, we studied the impact of a heparin supplement on MSC procoagulative properties. However, MSCs still retained procoagulant activity toward blood from children receiving a therapeutic dose of unfractionated heparin. An analysis of the mechanisms responsible for the procoagulant effect of MSCs/EVs revealed the presence of tissue factor and other proteins involved in coagulation-associated pathways. Also, we found that some MSCs and EVs were positive for annexin V, which implies the presence of phosphatidylserine on their surfaces, which can potentiate clot formation. Thus, we revealed procoagulant activity of MSCs/EVs associated with the presence of phosphatidylserine and tissue factor, which requires further analysis to avoid adverse effects of MSC therapy in patients with a risk of thrombosis.

2007 ◽  
Vol 92 (11) ◽  
pp. 4352-4358 ◽  
Author(s):  
Guenther Boden ◽  
Vijender R. Vaidyula ◽  
Carol Homko ◽  
Peter Cheung ◽  
A. Koneti Rao

Abstract Context: Type 2 diabetes mellitus (T2DM) is a hypercoagulable state. Tissue factor (TF) is the principal initiator of blood coagulation. Objective: Our objective was to examine the effects of hyperglycemia and hyperinsulinemia on the TF pathway of blood coagulation in T2DM. Design: Three study protocols were used: 1) acute correction of hyperglycemia (with iv insulin) followed by 24 h of euglycemia, 2) 24 h of selective hyperinsulinemia, and 3) 24 h of combined hyperinsulinemia and hyperglycemia. Setting: The study took place at a clinical research center. Study Participants: Participants included 18 T2DM patients and 22 nondiabetic controls. Results: Basal TF-procoagulant activity (TF-PCA), monocyte TF mRNA, plasma coagulation factor VII (FVIIc), and thrombin-anti-thrombin complexes were higher in T2DM than in nondiabetic controls, indicating a chronic procoagulant state. Acutely normalizing hyperglycemia over 2–4 h resulted in a small (∼7%) but significant decline in TF-PCA with no further decline over 24 h. Raising insulin levels alone raised TF-PCA by 30%, whereas raising insulin and glucose levels together increased TF-PCA (by 80%), thrombin-anti-thrombin complexes, and prothrombin fragment 1.2. Plasma FVIIa and FVIIc declined with increases in TF-PCA. Conclusion: We conclude that the combination of hyperglycemia and hyperinsulinemia, common in poorly controlled patients with T2DM, contributes to a procoagulant state that may predispose these patients to acute cardiovascular events.


Blood ◽  
2006 ◽  
Vol 107 (3) ◽  
pp. 980-986 ◽  
Author(s):  
Xin Huang ◽  
Wei-Qun Ding ◽  
Joshua L. Vaught ◽  
Roman F. Wolf ◽  
James H. Morrissey ◽  
...  

AbstractTissue factor (TF) initiates blood coagulation, but its expression in the vascular space requires a finite period of time. We hypothesized that targeting exogenous tissue factor to sites of vascular injury could lead to accelerated hemostasis. Since phosphatidylserine (PS) is exposed on activated cells at sites of vascular injury, we cloned the cDNA for a chimeric protein consisting of the extracellular domain of TF (called soluble TF or sTF) and annexin V, a human PS-binding protein. Both the sTF and annexin V domains had ligand-binding activities consistent with their native counterparts, and the chimera accelerated factor X activation by factor VIIa. The chimera exhibited biphasic effects upon blood coagulation. At low concentrations it accelerated blood coagulation, while at higher concentrations it acted as an anticoagulant. The chimera accelerated coagulation in the presence of either unfractionated or low-molecular-weight heparins more potently than factor VIIa and shortened the bleeding time of mice treated with enoxaparin. The sTF-annexin V chimera is a targeted procoagulant protein that may be useful in accelerating thrombin generation where PS is exposed to the vasculature, such as may occur at sites of vascular injury or within the vasculature of tumors.


2019 ◽  
Vol 70 (1) ◽  
pp. e661
Author(s):  
Mattias Mandorfer ◽  
Johannes Thaler ◽  
Lena Hell ◽  
Philipp Schwabl ◽  
Lukas Wisgrill ◽  
...  

Blood ◽  
1973 ◽  
Vol 41 (5) ◽  
pp. 671-678 ◽  
Author(s):  
Leo R. Zacharski ◽  
Leon W. Hoyer ◽  
O. Ross McIntyre

Abstract Immunologic methods were employed in an attempt to identify a potent procoagulant present in homogenates of human skin fibroblasts cultured in vitro. The activity of this procoagulant was restricted to the early stages of coagulation and was heretofore considered to be due to tissue factor (tissue thromboplastin, factor III) either alone or in combination with one or more of the first-stage coagulation factors (VIII, IX, XI, XII). The present studies demonstrated that procoagulant activity was not diminished by incubation with anti-VIII or anti-IX antibodies of human origin or with anti-VIII antibody of rabbit origin. Furthermore, cell culture homogenates failed to bind antifactor VIII antibody and did not contain an inhibitor of the reaction between factor VIII and its antibody. By contrast, procoagulant activity was obliterated by an antibody to tissue factor protein regardless of whether plasmas deficient in factor VIII, IX, XI, or XII were used in the assay system. The antitissue factor antibody failed to block the procoagulant effect after tissue factor had complexed factor VII. The procoagulant, therefore, consisted entirely of tissue factor.


PLoS ONE ◽  
2020 ◽  
Vol 15 (10) ◽  
pp. e0240189
Author(s):  
Marcela Rosas ◽  
David A. Slatter ◽  
Samya G. Obaji ◽  
Jason P. Webber ◽  
Jorge Alvarez-Jarreta ◽  
...  

1998 ◽  
Vol 79 (05) ◽  
pp. 1025-1028 ◽  
Author(s):  
Ling Zhou ◽  
Patrick Stordeur ◽  
Aurore de Lavareille ◽  
Kris Thielemans ◽  
Paul Capel ◽  
...  

SummaryThe CD40 molecule expressed on endothelial cells has been shown to transduce activation signals resulting in upregulation of adhesion molecules. Herein, we studied the impact of CD40 engagement on the induction of tissue factor (TF)-dependent procoagulant activity (PCA) at the surface of human umbilical vein endothelial cells (HUVECs). First, we found that co-incubation of HUVECs with 3T6 fibroblasts transfected with the CD40L gene (3T6-CD40L) resulted in a clear induction of PCA which was not observed with control untransfected fibroblasts. The specificity of this finding was established by inhibition experiments using monoclonal antibodies (mAbs) blocking CD40 or CD40L. PCA induced by CD40 ligation was TF-related as it was not observed in factor VII-deficient plasma and was associated with the accumulation of TF mRNA. To investigate the role of CD40/CD40L interactions in the induction of endothelial cell PCA by lymphocytes, interferon (IFN)-γ-stimulated EC were incubated with T cells in the absence or presence of anti-CD40 or anti-CD40L mAb. The 60-70% inhibition of PCA induced by these mAbs but not their isotype-matched control indicated that the CD40 pathway is involved in the induction of PCA resulting from interactions between activated HUVECs and T cells. We conclude that activation signals elicited by CD40 engagement on endothelial cells result in the induction of TF-dependent PCA. The CD40/CD40L pathway might therefore be involved in the development of prothrombic states during diseases associated with endothelial cell and T cell activation.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3736-3736
Author(s):  
Anna Falanga ◽  
Alfonso Vignoli ◽  
Marina Marchetti ◽  
Laura Russo ◽  
Marina Panova-Noeva ◽  
...  

Abstract Clinical data suggest an increased thrombotic risk in patients with ET or PV carrying the JAK2V617F mutation. Laboratory data from our group show that ET patients carrying the JAK2V617F mutation are characterized by an enhanced platelet and neutrophil activation status (Falanga et al, Exp Hem 2007) and blood coagulation activation (Marchetti et al, Blood 2008), as compared to JAK2 wild-type ET. Since monocytes significantly contribute to blood coagulation activation as an important source of circulating tissue factor (TF), in this study we aimed to characterize the prothrombotic phenotype of monocytes from ET and PV patients and to evaluate whether and to what extent it is influenced by the JAK2V617F mutation. Twenty-four ET patients (10 JAK2 wild-type; 14 JAK2V617F carriers with 2%–35% mutant allele burden), 27 PV patients (all JAK2V617F carriers, 16 with 9%– 44% and 11 with 60%–100% allele burden, respectively), and 20 age-matched healthy subjects (controls, C) were enrolled into the study. Monocyte-associated TF antigen was measured on the cell surface by whole blood flow-cytometry, in both basal condition and after in vitro stimulation by bacterial endotoxin (lypopolysaccharide, LPS), as well as in cell lysates by ELISA. Monocyte procoagulant activity was evaluated by the Calibrated Automated Thrombogram (CAT) as the capacity of isolated monocyte lysates to induce thrombin generation in normal pool plasma. In basal conditions, significantly (p<0.05) higher surface levels of TF were measured on monocytes from ET (17.1±3.2% positive cells) and PV (24.4±3.7% positive cells) patients compared to C (8.2±1.9% positive cells). Similarly, the total TF antigen content of cell lysates was significantly increased in patients compared to C. The analysis of the data according to JAK2V617F mutational status, showed a gradient of increased TF expression starting from JAK2V617F negative patients (11.7±2.5%), versus JAK2V617F ET and PV subjects with <50% allele burden (20.3±3.6% and 23.2±2.8%, respectively), versus JAK2V617F PV patients with >50% allele burden (26.1±4.2%). The in vitro LPS stimulation significantly increased TF expression on monocytes from all study subjects and C compared to non-stimulated monocytes (p<0.05 for all groups), with a more elevated expression by monocytes from PV and ET patients compared to C. However, the relative increase in TF expression was greater in C (=3.7 fold) compared to both ET (=2.2 fold) and PV (=2 fold) patients. As observed in basal conditions, LPS-induced TF was higher in JAK2V617F positive patients as compared to negative, with the highest expression in JAK2V617F PV carriers with >50% allele load. Thrombin generation induced by monocytes from ET and PV patients was significantly increased compared to controls, as determined by significantly higher thrombin peaks (ET=145±12, PV=142±17, C=72.2±5 nM), and quantity of thrombin generated in time, i.e. the endogenous thrombin potential (ETP) (ET=1143±34, PV=1074±64, C=787±58 nM*min). The JAK2V617F PV subjects with >50% allele burden presented with the highest thrombin generation capacity (peak= 184±34 nM; ETP= 1268±32 nM). Our data indicate that the expression of the JAK2V617F mutation in ET and PV patients may confer to monocytes a different hemostatic phenotype in terms of increased expression of surface TF and thrombin generation capacity. These findings are in agreement with the previous observation of a hypercoagulable state associated with this mutation and suggest a new mechanism linking hemostatic cellular phenotypic alteration to genetic dysfunction in patients with myeloproliferative disease.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4975-4975
Author(s):  
Venencia Albert ◽  
Arulselvi Subramanian ◽  
Hara Prasad Pati

Abstract Introduction Microparticles (MPs) are small membrane blebs that are released in response to cellular activation or apoptosis from the cell surface by proteolytic cleavage of the cytoskeleton. Previous studies have reported low levels of naturally produced cell-derived microparticle in healthy individuals. This study was conducted to assess whether circulating microparticles induce thrombin generation leading to low grade activation of the coagulation system in healthy individuals. Materials & Methods Flow cytometry analysis of three phenotypes of Platelet derived (PMP), Endothelial derived (EMP) and TF bearing (TFMP) microparticles was done in in the 20 healthy individuals. Triple gating strategy (i) particle size(<0.5µm) (ii) expression of cell surface markers (PMP, CD42a+; EMP, CD62E+; TF bearing MP(TFMP), CD142+) and (iii) phosphatidylserine(Annexin V+) was used. Plasma concentrations of thrombomodulin (TM), Tissue Factor (TF), Tissue factor pathway inhibitor (TFPI), Protein C (PC), Free Protein S (PS), Thrombin antithrombin (TAT) complexes, soluble fibrin monomer (sFM) was done assess the status of coagulation system. Spearman's rank correlation was done examine the role of cell-derived microparticles as critical effectors of thrombosis. Results: the study group comprised of 75% males of the age group (mean±SD27.3±4.32. Total Microparticles (Annexin V+) enumerated in plasma was 1125±355 (count/µl). 52.8% were TFMP [median (IQR)] [380.3(301.8-710.1) (count/µl)]. Highest number of MP originated from activated platelets [249.1(198.9-404.5) (count/µl)], followed by endothelial cells [140.9(124.9-286.0) (count/µl)]. A statistically significant and inverse correlation of EMP's with TAT complex was observed [12.7(6.8-20.1) ng/ml] (r = -0.46, p = 0.01), PS (93.4±20.6 %) (r = -0.52, p = 0.01) and TM (11.4±4.47 ng/ml) (r = -0.56, p = 0.008). Also, TFMP significantly correlated with TF [3.9(3.0-5.0)] (r = 0.46, p = 0.03) and PC (90.4±17.7) (r=-0.42, p=0.04). No correlation was observed between PMP numbers and coagulation pathway markers. Conclusion: Sub optimal degree of coagulation and natural anticoagulation pathway are occurring in healthy individuals. Microparticles moderately correlated with TF levels. Low EMP's levels correlated with the anticoagulation markers and thrombin generation, suggesting that EMPS may have a role in inhibition of thrombosis. The impact of endothelial vesiculation on basal coagulation should be studied further. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4788-4788
Author(s):  
Damien DG Gheldof ◽  
François Mullier ◽  
Bernard Chatelain ◽  
jean-Michel Dogne ◽  
Christian Chatelain

Abstract Abstract 4788 Introduction: Thrombosis is a common complication of patients with malignancies. Patients with hematological malignancy have a 28 fold increase risk to develop venous thromboembolism (VTE). A population-based cohort was used to determine the incidence and risk factors associated with development of venous thromboembolism (VTE) among Californians diagnosed with acute leukemia between 1993 to 1999. Principal outcomes were deep vein thrombosis in the lower and upper extremities, pulmonary embolism, and mortality. Among 5,394 cases with acute myelogenous leukemia (AML), the 2-year cumulative incidence of VTE was 281 (5.2%). Sixty-four% of the VTE events occurred within 3 months of AML diagnosis. The induction of hypercoagulable state mechanisms is not fully understood to date. Multifactorial aspects such as physic immobility, chemotherapy adverse effects or the overexpression of several procoagulant substances (cytokines, cysteine protease and tissue factor) by cancer cells are often provoked. Several studies strongly suggest that microvesicles (MVs) harboring tissue factor activity may have a primary role in VTE. MVs are small membrane vesicles shed from normal and/or tumor cells following activation or apoptosis. MVs may present TF and negatively charged phospholipids (PL) such as phosphatidylserine on their membrane. These elements are thought to be implicated in the procoagulant activity (PCA). Objectives: The aim of this study was to assess the capacity of untreated acute promyelocytic leukemia cells to shed procoagulant MVs. Methods: Acute promyelocytic leukemia (APL) cells lines (NB4 and HL-60) were cultured 48h in medium at 600,000 cells/mL. Cells and MVs were separated by filtrations (0.1–0.22–0.45–0.65μm). The PCA was assessed by thrombin generation assay. Alternatively, MVs were incubated with anti-TF antibodies (10μg/mL) or with annexin V (0,5μM to assess the contribution of TF and phospholipids to the PCA. Moreover HL-60 cells were incubated with HgCl2 which promote di-S bond formation (activation of TF). Results: NB4 cells and HL-60 cells can stimulate thrombin generation. HL60 cells reduced the lagtime 3.9-fold and increased the peak 1.6-fold in comparison to CTL and NB4 cells decreased the lagtime 10.9-fold and increased the peak 6.7-fold in comparison to CTL. No PCA was observed in HL-60 filtered with 0.65 μm membrane (no statistical difference in lagtime peak and ETP). By contrast, NB4 cells can support thrombin generation activity when filtered at different sizes. MVs of sizes <0.65 and <0.45 μm decreased the lagtime 4.2- and 3.8-fold, respectively and increased the peak of thrombin 4.6- and 4.1-fold, respectively. MVs of sizes lower than 0.22 and <0.1 μm reduced the lagtime 2.4- and 1.6-fold, respectively and increased the peak 2.3- and 1.4-fold, respectively. Thrombin generation activity of MVs of size <0.65 μm derived from NB4 cells wass abolished when anti-TF antibodies or annexin V were preincubated Discussion: NB4 cells and HL-60 cells have different PCA. NB4 cells have a higher procoagulant activity and most of the PCA is linked to MVs of size under 0.45 μm. NB4 cells spontaneously release different sized MVs which can support thrombin generation. By using an anti-TF function-blocking antibody (HTF-1) and annexin V which binds phosphatidylserine, we confirmed that the PCA of MVs is related to the expression of active TF and PL. HL-60 cells have a weaker procoagulant activity because TF is mostly present in an inactive form (activation of TF by reduction agent such as HgCl2 increased the PCA of HL-60 cells of +/− 35%). Moreover HL-60 cells do not produce MVs<0.65 μm associated with PCA. Conclusions: APL NB4 cells and HL-60 cells can stimulate thrombin generation. NB4 cells release MVs (of size <0.65 μm) whose procoagulant activity is mediated by TF and PL. These MVs could have a prognostic value for VTE in patient with APL. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document