15 research outputs found

    Genetic and Pharmacological Modifications of Thrombin Formation in Apolipoprotein E-deficient Mice Determine Atherosclerosis Severity and Atherothrombosis Onset in a Neutrophil-Dependent Manner

    Get PDF
    Background: Variations in the blood coagulation activity, determined genetically or by medication, may alter atherosclerotic plaque progression, by influencing pleiotropic effects of coagulation proteases. Published experimental studies have yielded contradictory findings on the role of hypercoagulability in atherogenesis. We therefore sought to address this matter by extensively investigating the in vivo significance of genetic alterations and pharmacologic inhibition of thrombin formation for the onset and progression of atherosclerosis, and plaque phenotype determination. Methodology/principal findings: We generated transgenic atherosclerosis-prone mice with diminished coagulant or hypercoagulable phenotype and employed two distinct models of atherosclerosis. Gene-targeted 50% reduction in prothrombin (FII/WT:ApoE/)(FII^{−/WT}:ApoE^{−/−}) was remarkably effective in limiting disease compared to control ApoE/ApoE^{−/−} mice, associated with significant qualitative benefits, including diminished leukocyte infiltration, altered collagen and vascular smooth muscle cell content. Genetically-imposed hypercoagulability in TMPro/Pro:ApoE/TM^{Pro/Pro}:ApoE^{−/−} mice resulted in severe atherosclerosis, plaque vulnerability and spontaneous atherothrombosis. Hypercoagulability was associated with a pronounced neutrophilia, neutrophil hyper-reactivity, markedly increased oxidative stress, neutrophil intraplaque infiltration and apoptosis. Administration of either the synthetic specific thrombin inhibitor Dabigatran etexilate, or recombinant activated protein C (APC), counteracted the pro-inflammatory and pro-atherogenic phenotype of pro-thrombotic TMPro/Pro:ApoE/TM^{Pro/Pro}:ApoE^{−/−} mice. Conclusions/significance: We provide new evidence highlighting the importance of neutrophils in the coagulation-inflammation interplay during atherogenesis. Our findings reveal that thrombin-mediated proteolysis is an unexpectedly powerful determinant of atherosclerosis in multiple distinct settings. These studies suggest that selective anticoagulants employed to prevent thrombotic events may also be remarkably effective in clinically impeding the onset and progression of cardiovascular disease

    Rethinking the donor's role in strengthening health systems through sexual and reproductive health and rights partnerships: A qualitative analysis of Ugandan case studies

    No full text
    International donors continue to prefer vertical programming over systems strengthening despite the universal health agenda. This study explored Dutch policy and practice towards health systems within sexual and reproductive health and rights-focused partnerships between the Netherlands Ministry of Foreign Affairs and civil society, through a document analysis, 13 in-depth interviews and a stakeholder workshop. The findings revealed that partnerships supported the Ugandan health system in unstructured ways and had difficulties finding synergies. To ensure sustained outcomes and respond to the renewed urgency of strong health systems in the face of crises, donors should incorporate systems strengthening as an explicit goal

    Morphometrical analysis of periadventitial cuff-induced atherosclerosis in mice with genetically imposed alterations in blood coagulation potential.

    No full text
    <p>(A) Representative hematoxylin and eosin (H&E)-stained sections of carotid arteries of FII<sup>−/+</sup>:ApoE<sup>−/−</sup>, TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> and control ApoE<sup>−/−</sup> mice (top row). Necrotic core areas of the atherosclerotic lesions were identified and quantified by using toluidine blue (TB) staining (second and third row). (B, C) Whereas hypocoagulable mice were significantly protected against plaque progression (26.5±12.6*10<sup>3</sup> in FII<sup>−/+</sup>:ApoE<sup>−/−</sup> vs. 69.2±18.4*10<sup>3</sup> µm<sup>2</sup> in ApoE<sup>−/−</sup> control mice, n = 10 per group, p<0.0001), pro-thrombotic mice developed severe and occlusive atherosclerotic burden (146.4±52.7*10<sup>3</sup> in TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> vs. 53.9±27.0*10<sup>3</sup> µm<sup>2</sup> in ApoE<sup>−/−</sup> control mice, n = 10 per group, p = 0.0001). The degree of stenosis in TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> reached an average of 88.6±8.1% (vs. 62.2±16.1% in ApoE<sup>−/−</sup> mice, n = 10 per group, p = 0.0002), whereas it was substantially lower in FII<sup>−/+</sup>:ApoE<sup>−/−</sup> mice (36.8±11.9% vs. 64.9±9.6% in ApoE<sup>−/−</sup> mice, n = 10 per group, p<0.0001). (A, D) Pearson's chi-squared test (<i>χ</i><sup>2</sup>) detected a significant difference in the number of advanced atherosclerotic lesions (presence of fibrous cap atheromata <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0055784#pone.0055784-Virmani1" target="_blank">[54]</a>) formed between FII<sup>−/+</sup>:ApoE<sup>−/−</sup> (4 out of 10) and TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> mice (10 out of 10) (n = 10 per group, p = 0.0108). In fact, the necrotic area within the lesions of the hypercoagulable mice was significantly increased: 56.2±10.8% of the total plaque area, as compared to 29.0±17.7% in the control ApoE<sup>−/−</sup> group (n = 10 per group, p = 0.0024). (E) Hypocoagulable mice showed more stable advanced lesions, as indicated by the significantly thicker fibrous caps in comparison to ApoE<sup>−/−</sup> mice (n = 10 per group, p = 0.0081). (F) Intima/media ratio was significantly increased in TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> mice, whereas profoundly decreased in FII<sup>−/+</sup>:ApoE<sup>−/−</sup> mice. Of note, the average outer diameter of the common carotid artery is 0.36 mm <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0055784#pone.0055784-vonderThusen1" target="_blank">[21]</a>, thus suggesting that TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> atherosclerotic plaques undergo a dramatic outward remodeling as indicated in panel (G). *<i>p</i><0.05; **<i>p</i><0.01; ***<i>p</i><0.001. Error bars represent mean ± SD. Arrows indicate examples of positive staining/fibrous cap thickness. Abbreviations: H&E – hematoxylin and eosin; AL – advanced atherosclerotic lesion.</p

    Hypercoagulability induces oxidative stress in granulocytes within the bone marrow compartment.

    No full text
    <p>Granulocytes and monocytes cell fractions in the bone marrow were significantly increased in TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> as compared to ApoE<sup>−/−</sup> control mice after 8 weeks on a regular chow diet (Granulocytes: 26.3±3.6% vs. 22.9±3.4%; n = 12 per group, p = 0.0292)(Monocytes: 12.3±0.6% vs. 8.8±0.7%; n = 12 per group, p<0.0001) (A, B). The significant increase in monocytes can be explained by the higher relative numbers of Ly6C<sup>HIGH</sup> monocyte cells in TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> mice bone marrow (Ly6C<sup>HIGH</sup> cells: 9.4±1.3% vs. 6.3±0.8%; n = 12 per group, p = 0.0002) (C). Using DHR123 FACS analysis, we analyzed the amount of oxidative burst activity in granulocytes and monocytes in the bone marrow after PMA stimulation. The monocytes did not show any differences in DHR signal and thus ROS activity, whereas in the granulocytes of the TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> mice, a significant increase was observed in the DHR signal when compared to ApoE<sup>−/−</sup> mice, indicating enhanced oxidative stress upon PMA stimulation in the TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> granulocytes present in the bone marrow (D, E). *<i>p</i><0.05; **<i>p</i><0.01; ***<i>p</i><0.001. Error bars represent mean ± SD. Abbreviations: DHR123– Dihydrorhodamine 123; ROS – Reactive Oxygen Species; PMA - Phorbol 12-Myristate 13-Acetate.</p

    The effects of variations in coagulation potential on atherogenesis in a spontaneous atherosclerosis model at 35 weeks on a regular chow diet.

    No full text
    <p>(A) Top row represents images of the aortic arch and its main branches, stained with hematoxylin and eosin (H&E), used to analyze the extent of atherosclerotic plaque burden. To determine plaque phenotype characteristics, sections were stained against α-smooth muscle actin (vascular smooth muscle cell content – second row), MAC-2<sup>+</sup> (macrophage infiltration – third row), Ly-6G (neutrophil recruitment – fourth row) and with Sirius red (collagen – bottom row). (B) Hypocoagulability in FII<sup>−/+</sup>:ApoE<sup>−/−</sup> significantly attenuated atherosclerosis plaque development (90.6±35.1*10<sup>3</sup> µm<sup>2</sup> total plaque burden) when compared to normal ApoE<sup>−/−</sup> mice (160.6±65.9*10<sup>3</sup> µm<sup>2</sup>)(n = 10 per group, p = 0.0084). Total plaque area in TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> mice was established 389.1±158.4*10<sup>3</sup> vs. 187.0±35.1*10<sup>3</sup> µm<sup>2</sup> in the corresponding control ApoE<sup>−/−</sup> group (n = 10 per group, p = 0.0010). (C) TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> mice atherosclerotic plaques demonstrated a significant decrease in intimal vascular smooth muscle cell content (2.2±1.3% of plaque area) compared to ApoE<sup>−/−</sup> mice (8.7±2.9% of plaque area)(n = 10 per group, p = 0.0016). Recruitment of macrophages within the lesions did not differ between all experimental groups (D). Neutrophil infiltration was significantly diminished in the lesions of hypocoagulable FII<sup>−/+</sup>:ApoE<sup>−/−</sup> mice (n = 10 per group, p = 0.0092 vs. ApoE<sup>−/−</sup> mice), and substantially increased in the TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> intima (n = 10 per group, p = 0.0094 vs. ApoE<sup>−/−</sup> mice) (E). A similar trend was observed with regard to collagen deposition within the atherosclerotic plaques. In FII<sup>−/+</sup>:ApoE<sup>−/−</sup> mice, 29.3±3.6% of the plaque area stained collagen-positive (n = 10 per group, p = 0.0002 vs. ApoE<sup>−/−</sup> mice). In contrast, Sirius red staining showed only 4.1±3.0% positivity for collagen in the TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> lesions (n = 10 per group, p = 0.0002 vs. ApoE<sup>−/−</sup> mice) (F). By 35 weeks (established duration of the experiment), we recorded the following fatal events: 6 of 16 TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup>, 1 of 11 FII<sup>−/+</sup>:ApoE<sup>−/−</sup> and 0 of 20 ApoE<sup>−/−</sup> control mice. Dead mice were not included from the study analyses. The exact cause of death remained unclear. Kaplan-Meier analysis of the survival data comparing TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> vs. ApoE<sup>−/−</sup> mice, as determined by the Gehan-Breslow-Wilcoxon test, indicated that hypercoagulability is linked to significantly higher spontaneous mortality rates (p = 0.0165) (G). No significant difference was found between FII<sup>−/+</sup>:ApoE<sup>−/−</sup> and ApoE<sup>−/−</sup> control mice (p = 0.3173) (data not shown).*<i>p</i><0.05; **<i>p</i><0.01; ***<i>p</i><0.001. Error bars represent mean ± SD. Arrows indicate examples of positive staining. Abbreviations: H&E – hematoxylin and eosin; α-SMA - α-smooth muscle actin; SR – Sirius red.</p

    Hypercoagulable TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> mice – a new mouse model of atherosclerotic plaque vulnerability.

    No full text
    <p>Here we present a new hypercoagulable atherosclerosis model, which closely mimics the composition and events leading to plaque destabilization, as normally observed in human atherothrombosis. In a series of sections, demonstrating carotid atherosclerotic plaques, obtained from TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> mice at 6 weeks after collar placement on high-fat diet regimen, we show multiple signs of plaque vulnerability. (A) A non-occlusive but rapidly progressing atherosclerotic lesion, characterized by abundant infiltration of leukocytes. (B) TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> mice plaques tend to rupture and dissect (upper arrow) even during the non-occlusive phase, accompanied by “silent” intraluminal thrombosis (lower arrows). Despite the detrimental pathologic characteristics of those lesions, these data confirm the hypothesis that arterial thrombosis might exist long before a fatal event takes place. This is further consolidated by the presence of so called “buried fibrous caps” (indicated by the arrows) in TM<sup>Pro/Pro</sup>:ApoE<sup>−/−</sup> mice plaques <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0055784#pone.0055784-Jackson1" target="_blank">[55]</a>, considered a marker of healed plaque ruptures, and also observed in human atherosclerosis. Blue color denotes a massive intraplaque hemorrhage (iron ions deposition) (C). Hypercoagulability induces a severe inflammatory and pro-necrotic intraplaque environment, leading to the formation of enormous necrotic core, thin fibrous caps, further plaque destabilization (D) and atherothrombosis (occlusive intraluminal thrombosis/abundant fibrin(ogen) deposition (indicated by the arrows)) (E). Thrombi undergo fibrotic organization involving vascular smooth muscle cells and fibroblasts ingrowth, and are then partially recanalized by newly formed vessels (arrows, blue color – iron deposition/presence of erythrocytes)(F).</p
    corecore