Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
J Thromb Haemost ; 15(3): 526-537, 2017 03.
Article in English | MEDLINE | ID: mdl-27992950

ABSTRACT

Essentials Methods were developed to image the hemostatic response in mouse femoral arteries in real time. Penetrating injuries produced thrombi consisting primarily of platelets. Similar to arterioles, a core-shell architecture of platelet activation occurs in the femoral artery. Differences from arterioles included slower platelet activation and reduced thrombin dependence. SUMMARY: Background Intravital studies performed in the mouse microcirculation show that hemostatic thrombi formed after penetrating injuries develop a characteristic architecture in which a core of fully activated, densely packed platelets is overlaid with a shell of less activated platelets. Objective Large differences in hemodynamics and vessel wall biology distinguish arteries from arterioles. Here we asked whether these differences affect the hemostatic response and alter the impact of anticoagulants and antiplatelet agents. Methods Approaches previously developed for intravital imaging in the mouse microcirculation were adapted to the femoral artery, enabling real-time fluorescence imaging despite the markedly thicker vessel wall. Results Arterial thrombi initiated by penetrating injuries developed the core-and-shell architecture previously observed in the microcirculation. However, although platelet accumulation was greater in arterial thrombi, the kinetics of platelet activation were slower. Inhibiting platelet ADP P2Y12 receptors destabilized the shell and reduced thrombus size without affecting the core. Inhibiting thrombin with hirudin suppressed fibrin accumulation, but had little impact on thrombus size. Removing the platelet collagen receptor, glycoprotein VI, had no effect. Conclusions These results (i) demonstrate the feasibility of performing high-speed fluorescence imaging in larger vessels and (ii) highlight differences as well as similarities in the hemostatic response in the macro- and microcirculation. Similarities include the overall core-and-shell architecture. Differences include the slower kinetics of platelet activation and a smaller contribution from thrombin, which may be due in part to the greater thickness of the arterial wall and the correspondingly greater separation of tissue factor from the vessel lumen.


Subject(s)
Femoral Artery/diagnostic imaging , Hemostasis , Microcirculation , Wounds, Penetrating/therapy , Adenosine Diphosphate/metabolism , Animals , Anticoagulants/pharmacology , Arterioles/metabolism , Blood Coagulation/drug effects , Blood Platelets/metabolism , Femoral Artery/injuries , Fibrin/metabolism , Hemodynamics , Intravital Microscopy , Mice , Mice, Inbred C57BL , Platelet Activation , Platelet Aggregation Inhibitors/pharmacology , Signal Transduction , Thrombin/antagonists & inhibitors , Thrombin/metabolism , Thromboplastin/metabolism , Thrombosis/diagnostic imaging , Thrombosis/drug therapy
2.
Cell Death Dis ; 6: e1706, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25811803

ABSTRACT

Ku70 (Lupus Ku autoantigen p70) is essential in nonhomologous end joining DNA double-strand break repair, and ku70(-/-) mice age prematurely because of increased genomic instability and DNA damage responses. Previously, we found that Ku70 also inhibits Bax, a key mediator of apoptosis. We hypothesized that Bax-mediated apoptosis would be enhanced in the absence of Ku70 and contribute to premature death observed in ku70(-/-) mice. Here, we show that ku70(-/-) bax(+/-) and ku70(-/-) bax(-/-) mice have better survival, especially in females, than ku70(-/-) mice, even though Bax deficiency did not decrease the incidence of lymphoma observed in a Ku70-null background. Moreover, we found that ku70(-/-) mice develop lung diseases, like emphysema and pulmonary arterial (PA) occlusion, by 3 months of age. These lung abnormalities can trigger secondary health problems such as heart failure that may account for the poor survival of ku70(-/-) mice. Importantly, Bax deficiency appeared to delay the development of emphysema. This study suggests that enhanced Bax activity exacerbates the negative impact of Ku70 deletion. Furthermore, the underlying mechanisms of emphysema and pulmonary hypertension due to PA occlusion are not well understood, and therefore ku70(-/-) and Bax-deficient ku70(-/-) mice may be useful models to study these diseases.


Subject(s)
Antigens, Nuclear/genetics , DNA-Binding Proteins/genetics , Lung Diseases/genetics , Lymphoma/genetics , bcl-2-Associated X Protein/genetics , Animals , Apoptosis/genetics , Disease Models, Animal , Female , Heart Diseases/genetics , Heart Diseases/pathology , Humans , Ku Autoantigen , Lung Diseases/pathology , Lymphoma/pathology , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...