38 research outputs found

    Concentration-dependent inhibition of ADP-induced P-selectin expression using 2-MeSAMP and TGX-221.

    No full text
    <p>Heparinized human whole blood (nā€Š=ā€Š4) was treated for 5 minutes at 37Ā°C with (A) PBS as control, 2-MeSAMP (10 and 100 ĀµM; ā€œ2-MeSā€) or a combination (100 ĀµM each) of 2-MeSAMP and MRS2179 and (B) propylene glycol (ā€œPGā€) as control or TGX-221 (0.5 or 2.2 ĀµM; ā€œTGXā€). Afterwards platelets were activated using ADP (final concentration: 20 ĀµM) and the percentage of P-selectin expressing platelets under a pre-set histogram marker was analyzed in flow cytometry using an anti-P-selectin mAb. Data are given as means and SD.</p

    P<sub>2</sub>Y<sub>12</sub>/P<sub>2</sub>Y<sub>1</sub> blockade in combination with TGX-221 prevents upregulation of the Mac-1 receptor on granulocytes.

    No full text
    <p>Granulocyte activation was measured before (ā€œbefore circ.ā€) and 30 minutes after hypothermic ECC in groups treated with PBS (control group), 2-MeSAMP and MRS2179 (ā€œP<sub>2</sub>Y blockā€; 100 ĀµM each), propylene glycol (ā€œPGā€, control group), TGX-221 (ā€œTGXā€; 2.2 ĀµM) and a combination of P<sub>2</sub>Y block (100 ĀµM each) and PI3K p110Ī² inhibition with TGX-221 (2.2 ĀµM; ā€œP<sub>2</sub>Y block + TGXā€;). Mac-1 expression on granulocytes was evaluated using geometric mean values of antibody fluorescence in flow cytometry. Data are given as means (nā€Š=ā€Š6) and SD; groups were compared using RM-ANOVA with Bonferroniā€™s multiple comparison test; *p<0.05; ***p<0.001.</p

    Blockade of P<sub>2</sub>Y<sub>12</sub> and P<sub>2</sub>Y<sub>1</sub> as well as PI3K p110Ī² inhibition profoundly inhibits hypothermic ECC-induced P-selectin expression on platelets and platelet microparticles.

    No full text
    <p>Prior to (ā€œbefore circ.ā€) and after hypothermic ECC (28Ā°C, 30 minutes) flow cytometric analysis of P-selectin expression on platelets and PMPs was performed in groups treated with either PBS as control, 2-MeSAMP and MRS2179 (100 ĀµM each; ā€œP<sub>2</sub>Y blockā€), propylene glycol (ā€œPGā€) as control, TGX-221 to inhibit PI3K p110Ī² (2.2 ĀµM; ā€œTGXā€) or a combination of 2-MeSAMP and MRS2179 (100 ĀµM each) as well as TGX-221 (2.2 ĀµM; ā€œP<sub>2</sub>Y block + TGXā€). Representative dot plot indicating the identification of PMPs, single platelets and aggregates according to their size and granularity (A). Representative histogram overlay including a marker to identify percentages of P-selectin expressing platelets before circulation without additional stimulation (grey filled), before circulation with addition of ADP (20 ĀµM; grey solid line) as well as 30 minutes after hypothermic ECC (black solid line) (B). Percentages of platelets (C) and PMPs (D) expressing P-selectin are depicted. Data in (C) and (D) are given as means (nā€Š=ā€Š6) and SD; groups were compared using RM-ANOVA with Bonferroniā€™s multiple comparison test; *p<0.05; **p<0.01, ***p<0.001.</p

    Effects of hypothermic ECC and platelet inhibitor treatment on platelet adhesion to the ECC surface, platelet-granulocyte aggregate formation and platelet counts.

    No full text
    <p>Human blood samples were either left untreated or treated with PBS (control group), a combination of 2-MeSAMP and MRS2179 (100 ĀµM each; ā€œP<sub>2</sub>Y blockā€), propylene glycol (ā€œPGā€, control group), TGX-221 (2.2 ĀµM; ā€œTGXā€) or a combination of 2-MeSAMP, MRS2179 and TGX-221 (ā€œP<sub>2</sub>Y block + TGXā€). Blood, which was treated accordingly, was circulated in an <i>ex vivo</i> ECC model at 28Ā°C for 30 minutes. A specific ELISA method was employed to detect platelet adhesion to the ECC surface (A; nā€Š=ā€Š6). Platelet-granulocyte aggregate formation was measured in flow cytometry before circulation (ā€œbefore circ.ā€) and after circulation in all treatment groups (B; nā€Š=ā€Š4) according to the fluorescence of an anti-CD15-PE antibody on aggregates (aggregate region in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0038455#pone-0038455-g003" target="_blank">figure 3A</a>) under a pre-set histogram marker. Platelet counts were measured in all samples (C; nā€Š=ā€Š6). Data are given as means and SD; normally distributed data were compared using RM-ANOVA with Bonferroniā€™s multiple comparison test (A, B); not normally distributed data were analyzed using a non-parametrical test (Friedman test with DunĒ¹s multiple comparison test; C); *p<0.05; **p<0.01; ***p<0.001.</p

    GPIIb/IIIa and GPIbĪ± expression as well as vWF binding are not influenced by <i>ex vivo</i> hypothermic ECC and antiplatelet agents, while platelet PI3K p110Ī² inhibition alone or in combination with P<sub>2</sub>Y<sub>12</sub>/P<sub>2</sub>Y<sub>1</sub> receptor blockade reduce GPIIb/IIIa activation induced by hypothermic ECC.

    No full text
    <p>Human blood was left untreated (ā€œbefore circ.ā€) or treated <i>ex vivo</i> with PBS (control), a combination of 2-MeSAMP and MRS2179 to block P<sub>2</sub>Y receptors (100 ĀµM each; ā€œP<sub>2</sub>Y blockā€), propylene glycol (ā€œPGā€, control), TGX-221 to inhibit PI3K p110Ī² (2.2 ĀµM; ā€œTGXā€) or a combination of 2-MeSAMP, MRS2179 (100 ĀµM each) and TGX-221 (2.2 ĀµM; ā€œP<sub>2</sub>Y block + TGXā€). All treated samples were circulated in an ECC model for 30 minutes at 28Ā°C. Expression of GPIIb (A; nā€Š=ā€Š4), activated GPIIb/IIIa (B; nā€Š=ā€Š6), GPIbĪ± (C; nā€Š=ā€Š6) as well as vWF binding (D; nā€Š=ā€Š4) were evaluated in flow cytometry using specific antibodies. ADP stimulation (20 ĀµM) before and after hypothermic ECC was performed as positive control for GPIbĪ± expression (C) and vWF binding (D) in the PBS group. Geometric mean fluorescence values of fluorescently labeled antibodies are given in diagrams as means and SD; not normally distributed data were analyzed using a non-parametrical test (Friedman test with DunĒ¹s multiple comparison test; A, D); normally distributed data (B, C) were compared using RM-ANOVA with Bonferroniā€™s multiple comparison test; *p<0.05; **p<0.01; ***p<0.001.</p

    Overview of a pharmacological strategy for platelet protection during hypothermic ECC employing P<sub>2</sub>Y<sub>12</sub> and P<sub>2</sub>Y<sub>1</sub> receptor blockers as well as a PI3K p110Ī² inhibitor.

    No full text
    <p>Overview of a pharmacological strategy for platelet protection during hypothermic ECC employing P<sub>2</sub>Y<sub>12</sub> and P<sub>2</sub>Y<sub>1</sub> receptor blockers as well as a PI3K p110Ī² inhibitor.</p

    Image_2_Crosstalk between cytotoxic CD8+ T cells and stressed cardiomyocytes triggers development of interstitial cardiac fibrosis in hypertensive mouse hearts.tiff

    No full text
    Graphical AbstractCardiac fibrosis developed from fibrotic remodelling due to hypertension leads to heart failure. Immune cells are abundantly accumulated in fibrotic regions, but exact mechanisms how they contributed to cardiac fibrosis are not clear. Here we demonstrated that CD8 T cells are major contributor to cardiac fibrosis in hypertensive hearts. Stressed cardiomyocytes express STING-dependent RAE-1 that activates NKG2D + CD8 T cells to induce apoptosis of stressed cardiomyocytes. Preventing STING signalling in stressed cardiomyocytes attenuates cardiac fibrosis. Pharmacologically inhibiting cardiomyocayte-RAE-1 and CD8+ T cell-NKG2D axis may be a potential therapeutic strategy to prevent cardiac fibrosis in HFpEF patients.</p

    Image_1_Crosstalk between cytotoxic CD8+ T cells and stressed cardiomyocytes triggers development of interstitial cardiac fibrosis in hypertensive mouse hearts.tiff

    No full text
    Graphical AbstractCardiac fibrosis developed from fibrotic remodelling due to hypertension leads to heart failure. Immune cells are abundantly accumulated in fibrotic regions, but exact mechanisms how they contributed to cardiac fibrosis are not clear. Here we demonstrated that CD8 T cells are major contributor to cardiac fibrosis in hypertensive hearts. Stressed cardiomyocytes express STING-dependent RAE-1 that activates NKG2D + CD8 T cells to induce apoptosis of stressed cardiomyocytes. Preventing STING signalling in stressed cardiomyocytes attenuates cardiac fibrosis. Pharmacologically inhibiting cardiomyocayte-RAE-1 and CD8+ T cell-NKG2D axis may be a potential therapeutic strategy to prevent cardiac fibrosis in HFpEF patients.</p

    Equal blocking of monocyte binding to endothelial cells by anti-EPCR and APC under flow conditions.

    No full text
    <p>Monocyte binding to HUVECs in dynamic adhesion assay after 5 min (left) and 10 min venous flow (middle), and after 1 min arterial flow (right). Pre-treatment of HUVECs with anti-EPCR (dark grey bars), anti-Mac-1 (light grey bars), or with activated protein C (drotrecogin alfa; crosshatched bars) diminished monocyte adhesion to an equal extent compared to control. (*** <i>p</i><0.0001; <sup>### </sup><i>p</i><0.0005; <sup>## </sup><i>p</i><0.005; <sup># </sup><i>p</i><0.05 vs. no blocking; ns ā€Š=ā€Š statistically not signifiant).</p

    <i>In vitro</i> Study of a Novel Stent Coating Using Modified CD39 Messenger RNA to Potentially Reduce Stent Angioplasty-Associated Complications

    Get PDF
    <div><p>Background</p><p>Stent angioplasty provides a minimally invasive treatment for atherosclerotic vessels. However, no treatment option for atherosclerosis-associated endothelial dysfunction, which is accompanied by a loss of CD39, is available, and hence, adverse effects like thromboembolism and restenosis may occur. Messenger RNA (mRNA)-based therapy represents a novel strategy, whereby <i>de novo</i> synthesis of a desired protein is achieved after delivery of a modified mRNA to the target cells.</p><p>Methods and Findings</p><p>Our study aimed to develop an innovative bioactive stent coating that induces overexpression of CD39 in the atherosclerotic vessel. Therefore, a modified CD39-encoding mRNA was produced by <i>in vitro</i> transcription. Different endothelial cells (ECs) were transfected with the mRNA, and CD39 expression and functionality were analyzed using various assays. Furthermore, CD39 mRNA was immobilized using poly(lactic-<i>co</i>-glycolic-acid) (PLGA), and the transfection efficiency in ECs was analyzed. Our data show that ECs successfully translate <i>in vitro</i>-generated CD39 mRNA after transfection. The overexpressed CD39 protein is highly functional in hydrolyzing ADP and in preventing platelet activation. Furthermore, PLGA-immobilized CD39 mRNA can be delivered to ECs without losing its functionality.</p><p>Summary</p><p>In summary, we present a novel and promising concept for a stent coating for the treatment of atherosclerotic blood vessels, whereby patients could be protected against angioplasty-associated complications.</p></div
    corecore