101
|
|
102
|
Abstract
BACKGROUND Venous thrombosis (VT) is an important cause of morbidity and mortality in clinical medicine. Animal models studying venous thrombosis are scarce and, in most cases, very crude and rely on sacrificing the animals to excise formed thrombi. Developing an in vivo murine model of venous thrombosis can be a powerful tool for studying venous thrombosis. OBJECTIVES We sought to use a high-frequency ultrasound system (HFUS) to dynamically and non-invasively monitor thrombus formation in the inferior vena cava (IVC) of mice. METHODS We developed a murine model of venous thrombosis using, for detection, the Vevo 770(®), a micro-imaging HFUS. Two different thrombosis models were used to generate thrombi in the IVC of C57Bl/6NCr mice: (i) ligation and (ii) application of ferric chloride (FeCl(3)). We then assessed venous thrombosis by HFUS. RESULTS In both models, measurements of the clot pathologically correlated favorably with measurements acquired with HFUS. Thrombus develops less than an hour after ligation or FeCl(3) -induced injury of the IVC and the size of the clot increases over time for up to 24 h. Importantly, we demonstrate that HFUS can be used to monitor the effect of an anticoagulant such as dalteparin until complete resolution of the thrombus. CONCLUSIONS These data show that HFUS assesses venous thrombosis in mice reliably and non-invasively. Developing a murine model of thrombosis using more accurate, and clinically more relevant, techniques such as ultrasonography, is a step towards a better understanding of the pathophysiology of venous thromboembolism.
Collapse
Affiliation(s)
- M N Aghourian
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
103
|
Luo W, Öhman M, Wright A, Kamrudin S, Wang H, Guo C, Eitzman D. Steatohepatitis and vascular thrombosis in apolipoprotein e deficient mice. Thromb Res 2012; 129:e166-7. [PMID: 22326191 DOI: 10.1016/j.thromres.2012.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 01/04/2012] [Accepted: 01/16/2012] [Indexed: 11/16/2022]
|
104
|
Kuijpers MJE, Heemskerk JWM. Intravital imaging of thrombus formation in small and large mouse arteries: experimentally induced vascular damage and plaque rupture in vivo. Methods Mol Biol 2012; 788:3-19. [PMID: 22130696 DOI: 10.1007/978-1-61779-307-3_1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Intravital fluorescence microscopy is increasingly used to measure experimental arterial thrombosis in large and small arteries of mice in vivo. This chapter describes protocols for applying this technology to detect and measure thrombi formed by: (1) ultrasound-induced rupture of an atherosclerotic plaque in the carotid artery of adult Apoe (-/-) mice; (2) FeCl(3) or ligation in the carotid artery of nonatherosclerotic mice; and (3) FeCl(3) in the mesenteric venules and arterioles of young mice. In addition, we describe a protocol using two-photon laser scanning microscopy for intraluminal scanning of thrombi formed in the carotid artery. These approaches provide important information that cannot be obtained with ex vivo methods and thus are likely to lead to new insights into the complex process of thrombosis.
Collapse
Affiliation(s)
- Marijke J E Kuijpers
- Department of Biochemistry, CARIM, Maastricht University, Maastricht, The Netherlands.
| | | |
Collapse
|
105
|
Thrombomodulin is required for the antithrombotic activity of thrombin mutant W215A/E217A in a mouse model of arterial thrombosis. Thromb Res 2011; 130:646-8. [PMID: 22178578 DOI: 10.1016/j.thromres.2011.11.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 11/04/2011] [Accepted: 11/15/2011] [Indexed: 01/31/2023]
Abstract
INTRODUCTION The thrombin mutant W215A/E217A (WE thrombin) has greatly reduced procoagulant activity, but it activates protein C in the presence of thrombomodulin and inhibits binding of platelet glycoprotein Ib to von Willebrand factor and collagen under flow conditions. Both thrombomodulin-dependent protein C activation and inhibition of platelet adhesion could contribute to the antithrombotic activity of WE thrombin. MATERIALS AND METHODS To assess the role of thrombomodulin, we administered WE thrombin to thrombomodulin-deficient (TM(Pro/Pro)) mice and measured the time to occlusive thrombus formation in the carotid artery after photochemical injury of the endothelium. RESULTS AND CONCLUSIONS Doses of WE thrombin ≥10μg/kg prolonged the thrombosis time of wild-type mice (>1.6-fold), while doses ≥100μg/kg only slightly prolonged the thrombosis time of TM(Pro/Pro) mice. We conclude that thrombomodulin plays a predominate role in mediating the antithrombotic effect of WE thrombin in the arterial circulation of mice after endothelial injury. Thrombomodulin-independent effects may occur only when high doses of WE thrombin are administered.
Collapse
|
106
|
La Bonte LR, Pavlov VI, Tan YS, Takahashi K, Takahashi M, Banda NK, Zou C, Fujita T, Stahl GL. Mannose-binding lectin-associated serine protease-1 is a significant contributor to coagulation in a murine model of occlusive thrombosis. THE JOURNAL OF IMMUNOLOGY 2011; 188:885-91. [PMID: 22156595 DOI: 10.4049/jimmunol.1102916] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Bleeding disorders and thrombotic complications constitute a major cause of death and disability worldwide. Although it is known that the complement and coagulation systems interact, no studies have investigated the specific role or mechanisms of lectin-mediated coagulation in vivo. FeCl(3) treatment resulted in intra-arterial occlusive thrombogenesis within 10 min in wild-type (WT) and C2/factor B-null mice. In contrast, mannose-binding lectin (MBL)-null and MBL-associated serine protease (MASP)-1/-3 knockout (KO) mice had significantly decreased FeCl(3)-induced thrombogenesis. Reconstitution with recombinant human (rh) MBL restored FeCl(3)-induced thrombogenesis in MBL-null mice to levels comparable to WT mice, suggesting a significant role of the MBL/MASP complex for in vivo coagulation. Additionally, whole blood aggregation demonstrated increased MBL/MASP complex-dependent platelet aggregation. In vitro, MBL/MASP complexes were captured on mannan-coated plates, and cleavage of a chromogenic thrombin substrate (S2238) was measured. We observed no significant differences in S2238 cleavage between WT, C2/factor B-null, MBL-A(-/-), or MBL-C(-/-) sera; however, MBL-null or MASP-1/-3 KO mouse sera demonstrated significantly decreased S2238 cleavage. rhMBL alone failed to cleave S2238, but cleavage was restored when rMASP-1 was added to either MASP-1/-3 KO sera or rhMBL. Taken together, these findings indicate that MBL/MASP complexes, and specifically MASP-1, play a key role in thrombus formation in vitro and in vivo.
Collapse
Affiliation(s)
- Laura R La Bonte
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
107
|
Rapidly regulating platelet activity in vivo with an antidote controlled platelet inhibitor. Mol Ther 2011; 20:391-7. [PMID: 22086230 DOI: 10.1038/mt.2011.226] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Millions of individuals are prescribed platelet inhibitors, such as aspirin and clopidogrel, to reduce their risk of thrombosis-related clinical events. Unfortunately many platelet inhibitors are contraindicated in surgical settings because of their inherent bleeding risk complicating the treatment of patients who require surgery. We describe the development of a potent antiplatelet agent, an RNA aptamer-termed Ch-9.14-T10 that binds von Willebrand factor (VWF) with high affinity and inhibits thrombosis in a murine carotid artery damage model. As expected, when this potent antiplatelet agent is administered, it greatly increases bleeding from animals that are surgically challenged. To improve this antiplatelet agent's safety profile, we describe the generation of antidotes that can rapidly reverse the activity of Ch-9.14-T10 and limit blood loss from surgically challenged animals. Our work represents the first antidote controllable antiplatelet agent, which could conceivably lead to improved medical management of patients requiring antiplatelet medication who also need surgery.
Collapse
|
108
|
Owens AP, Lu Y, Whinna HC, Gachet C, Fay WP, Mackman N. Towards a standardization of the murine ferric chloride-induced carotid arterial thrombosis model. J Thromb Haemost 2011; 9:1862-3. [PMID: 21884567 DOI: 10.1111/j.1538-7836.2011.04287.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- A P Owens
- Division of Hematology/Oncology Department of Medicine, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, NC, USA
| | | | | | | | | | | |
Collapse
|
109
|
Hechler B, Freund M, Alame G, Leguay C, Gaertner S, Cazenave JP, Petitou M, Gachet C. The antithrombotic activity of EP224283, a neutralizable dual factor Xa inhibitor/glycoprotein IIbIIIa antagonist, exceeds that of the coadministered parent compounds. J Pharmacol Exp Ther 2011; 338:412-20. [PMID: 21527535 DOI: 10.1124/jpet.111.181321] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
EP224283 combines in a single molecule idraparinux and tirofiban, which allows obtaining a predictable and sustained antiplatelet effect through the transfer of the pharmacokinetics properties of idraparinux to the anti-αIIbβ3 antagonist. The activity can be instantaneously neutralized by injection of avidin, a specific antidote. We have tested the effects of this new profile anticoagulant in various thrombosis models. The antithrombotic effect of EP224283 was compared with those of the parent compounds used alone or in association at doses achieving low to moderate inhibition of platelet aggregation ex vivo. In a model of systemic thromboembolism independent of thrombin generation, tirofiban and EP224283 had similar effects at equimolar doses. On the other hand, EP224283 was more potent than tirofiban or idraparinux under thrombin-dependent conditions. In a ferric chloride-induced thrombosis model, EP224283 was more potent than either parent compound or their combination. Similar results were obtained after atherosclerotic plaque rupture in ApoE(-/-) mice. Thus, the dual action of EP224283 exceeds that of the parent compounds used in combination. A possible explanation is that EP224283 could concentrate antithrombin inside the thrombus by binding to αIIbβ3 through the tirofiban moiety, as shown by immunolabeling of the occluded vessel. No prolongation of the bleeding time was observed at doses achieving strong antithrombotic effects, suggesting that low to moderate αIIbβ3 inhibition combined with factor Xa inhibition minimizes the bleeding risk. The favorable antithrombotic profile of EP224283 together with its possible neutralization by avidin makes it an interesting drug candidate for the treatment and prevention of acute ischemic events.
Collapse
Affiliation(s)
- Béatrice Hechler
- Unité Mixte de Recherche S949, Institut National de la Santé et de la Recherche Médicale, Université de Strasbourg, Etablissement Français du Sang-Alsace, 10 Rue Spielmann, BP 36, 67065 Strasbourg Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
110
|
Myerson J, He L, Lanza G, Tollefsen D, Wickline S. Thrombin-inhibiting perfluorocarbon nanoparticles provide a novel strategy for the treatment and magnetic resonance imaging of acute thrombosis. J Thromb Haemost 2011; 9:1292-300. [PMID: 21605330 PMCID: PMC3686484 DOI: 10.1111/j.1538-7836.2011.04339.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND As a regulator of the penultimate step in the coagulation cascade, thrombin represents a principal target of direct and specific anticoagulants. OBJECTIVE A potent thrombin inhibitor complexed with a colloidal nanoparticle was devised as a first-in-class anticoagulant with prolonged and highly localized therapeutic impact conferred by its multivalent thrombin-absorbing particle surface. METHODS PPACK (Phe[D]-Pro-Arg-Chloromethylketone) was secured covalently to the surface of perfluorocarbon-core nanoparticle structures. PPACK and PPACK nanoparticle inhibition of thrombin were assessed in vitro via thrombin activity against a chromogenic substrate. In vivo antithrombotic activity of PPACK, heparin, non-functionalized nanoparticles and PPACK nanoparticles was assessed through intravenous (i.v.) administration prior to acute photochemical injury of the common carotid artery. Perfluorocarbon particle retention in extracted carotid arteries from injured mice was assessed via (19) F magnetic resonance spectroscopy (MRS) and imaging (MRI) at 11.7 T. Activated partial thromboplastin time (APTT) measurements determined the systemic effects of the PPACK nanoparticles at various times after injection. RESULTS An optical assay verified that PPACK nanoparticles exceeded PPACK's intrinsic activity against thrombin. Application of an in vivo acute arterial thrombosis model demonstrated that PPACK nanoparticles outperformed both heparin (P=0.001) and uncomplexed PPACK (P = 0.0006) in inhibiting thrombosis. (19) F MRS confirmed that PPACK nanoparticles specifically bound to sites of acute thrombotic injury. APTT normalized within 20 min of PPACK nanoparticles injection. CONCLUSIONS PPACK nanoparticles present thrombin-inhibiting surfaces at sites of acutely forming thrombi that continue to manifest local clot inhibition even as systemic effects rapidly diminish and thus represent a new platform for localized control of acute thrombosis.
Collapse
Affiliation(s)
- J Myerson
- Washington University, Saint Louis, MO, USA
| | | | | | | | | |
Collapse
|
111
|
Xu Z, Kamocka M, Alber M, Rosen ED. Computational approaches to studying thrombus development. Arterioscler Thromb Vasc Biol 2011; 31:500-5. [PMID: 21325672 DOI: 10.1161/atvbaha.110.213397] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In addition to descriptive biological models, many computational models have been developed for hemostasis/thrombosis that provide quantitative characterization of thrombus development. Simulations using computational models that have been developed for coagulation reactions, platelet activation, and fibrinogen assembly have been shown to be in close agreement with experimental data. Models of processes involved in hemostasis/thrombosis are being integrated to simulate the development of the thrombus simultaneously in time and space. Further development of computational approaches can provide quantitative insights leading to predictions that are not obvious from qualitative biological models.
Collapse
Affiliation(s)
- Zhiliang Xu
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | | | | |
Collapse
|
112
|
Eckly A, Hechler B, Freund M, Zerr M, Cazenave JP, Lanza F, Mangin PH, Gachet C. Mechanisms underlying FeCl3-induced arterial thrombosis. J Thromb Haemost 2011; 9:779-89. [PMID: 21261806 DOI: 10.1111/j.1538-7836.2011.04218.x] [Citation(s) in RCA: 184] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND The FeCl(3)-induced vascular injury model is widely used to study thrombogenesis in vivo, but the processes leading to vascular injury and thrombosis are poorly defined. OBJECTIVES The aim of our study was to better characterize the mechanisms of FeCl(3)-induced vascular injury and thrombus formation, in order to evaluate the pathophysiological relevance of this model. METHODS FeCl(3) was applied at different concentrations (from 7.5% to 20%) and for different time periods (up to 5 min) to mouse carotid or mesenteric arteries. RESULTS Under all the conditions tested, ultrastructural analysis revealed that FeCl(3) diffused through the vessel wall, resulting in endothelial cell denudation without exposure of the inner layers. Hence, only the basement membrane components were exposed to circulating blood cells and might have contributed to thrombus formation. Shortly after FeCl(3) application, numerous ferric ion-filled spherical bodies appeared on the endothelial cells. Interestingly, platelets could adhere to these spheres and form aggregates. Immunogold labeling revealed important amounts of tissue factor at their surface, suggesting that these spheres may play a role in thrombin generation. In vitro experiments indicated that FeCl(3) altered the ability of adhesive proteins, including collagen, fibrinogen and von Willebrand factor, to support platelet adhesion. Finally, real-time intravital microscopy showed no protection against thrombosis in GPVI-immunodepleted and β(1)(-/-) mice, suggesting that GPVI and β(1) integrins, known to be involved in initial platelet adhesion and activation, do not play a critical role in FeCl(3)-induced thrombus formation. CONCLUSION This model should be used cautiously, in particular to study the earliest stage of thrombus formation.
Collapse
Affiliation(s)
- A Eckly
- UMR-S949 Inserm-Université de Strasbourg, Strasbourg, France
| | | | | | | | | | | | | | | |
Collapse
|
113
|
Cardenas JC, Owens AP, Krishnamurthy J, Sharpless NE, Whinna HC, Church FC. Overexpression of the cell cycle inhibitor p16INK4a promotes a prothrombotic phenotype following vascular injury in mice. Arterioscler Thromb Vasc Biol 2011; 31:827-33. [PMID: 21233453 PMCID: PMC3086817 DOI: 10.1161/atvbaha.110.221721] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2010] [Accepted: 01/03/2011] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Age-associated cellular senescence is thought to promote vascular dysfunction. p16(INK4a) is a cell cycle inhibitor that promotes senescence and is upregulated during normal aging. In this study, we examine the contribution of p16(INK4a) overexpression to venous thrombosis. METHODS AND RESULTS Mice overexpressing p16(INK4a) were studied with 4 different vascular injury models: (1) ferric chloride (FeCl(3)) and (2) Rose Bengal to induce saphenous vein thrombus formation; (3) FeCl(3) and vascular ligation to examine thrombus resolution; and (4) lipopolysaccharide administration to initiate inflammation-induced vascular dysfunction. p16(INK4a) transgenic mice had accelerated occlusion times (13.1 ± 0.4 minutes) compared with normal controls (19.7 ± 1.1 minutes) in the FeCl(3) model and 12.7 ± 2.0 and 18.6 ± 1.9 minutes, respectively in the Rose Bengal model. Moreover, overexpression of p16(INK4a) delayed thrombus resolution compared with normal controls. In response to lipopolysaccharide treatment, the p16(INK4a) transgenic mice showed enhanced thrombin generation in plasma-based calibrated automated thrombography assays. Finally, bone marrow transplantation studies suggested increased p16(INK4a) expression in hematopoietic cells contributes to thrombosis, demonstrating a role for p16(INK4a) expression in venous thrombosis. CONCLUSIONS Venous thrombosis is augmented by overexpression of the cellular senescence protein p16(INK4a).
Collapse
Affiliation(s)
- Jessica C Cardenas
- Department of Pathology and Laboratory Medicine, University of North Carolina-Chapel Hill, NC 27599-7035, USA
| | | | | | | | | | | |
Collapse
|
114
|
Heidt T, Deininger F, Peter K, Goldschmidt J, Pethe A, Hagemeyer CE, Neudorfer I, Zirlik A, Weber WA, Bode C, Meyer PT, Behe M, von Zur Mühlen C. Activated platelets in carotid artery thrombosis in mice can be selectively targeted with a radiolabeled single-chain antibody. PLoS One 2011; 6:e18446. [PMID: 21479193 PMCID: PMC3068185 DOI: 10.1371/journal.pone.0018446] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Accepted: 02/28/2011] [Indexed: 01/29/2023] Open
Abstract
Background Activated platelets can be found on the surface of inflamed, rupture-prone
and ruptured plaques as well as in intravascular thrombosis. They are key
players in thrombosis and atherosclerosis. In this study we describe the
construction of a radiolabeled single-chain antibody targeting the
LIBS-epitope of activated platelets to selectively depict platelet
activation and wall-adherent non-occlusive thrombosis in a mouse model with
nuclear imaging using in vitro and ex vivo
autoradiography as well as small animal SPECT-CT for in
vivo analysis. Methodology/Principal Findings LIBS as well as an unspecific control single-chain antibody were labeled with
111Indium (111In) via bifunctional DTPA
( = 111In-LIBS/111In-control).
Autoradiography after incubation with 111In-LIBS on activated
platelets in vitro (mean 3866±28 DLU/mm2,
4010±630 DLU/mm2 and 4520±293 DLU/mm2)
produced a significantly higher ligand uptake compared to
111In-control (2101±76 DLU/mm2, 1181±96
DLU/mm2 and 1866±246 DLU/mm2) indicating a
specific binding to activated platelets; P<0.05.
Applying these findings to an ex vivo mouse model of
carotid artery thrombosis revealed a significant increase in ligand uptake
after injection of 111In-LIBS in the presence of small thrombi
compared to the non-injured side, as confirmed by histology
(49630±10650 DLU/mm2 vs. 17390±7470
DLU/mm2; P<0.05). These findings could
also be reproduced in vivo. SPECT-CT analysis of the
injured carotid artery with 111In-LIBS resulted in a significant
increase of the target-to-background ratio compared to
111In-control (1.99±0.36 vs. 1.1±0.24;
P<0.01). Conclusions/Significance Nuclear imaging with 111In-LIBS allows the detection of platelet
activation in vitro and ex vivo with high
sensitivity. Using SPECT-CT, wall-adherent activated platelets in carotid
arteries could be depicted in vivo. These results encourage
further studies elucidating the role of activated platelets in plaque
pathology and atherosclerosis and might be of interest for further
developments towards clinical application.
Collapse
Affiliation(s)
- Timo Heidt
- Department of Cardiology and Angiology, University of Freiburg, Freiburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Bellido-Martín L, Chen V, Jasuja R, Furie B, Furie BC. Imaging fibrin formation and platelet and endothelial cell activation in vivo. Thromb Haemost 2011; 105:776-82. [PMID: 21437353 DOI: 10.1160/th10-12-0771] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2010] [Accepted: 02/04/2011] [Indexed: 11/05/2022]
Abstract
Over the past six decades research employing in vitro assays has identified enzymes, cofactors, cell receptors and associated ligands important to the haemostatic process and its regulation. These studies have greatly advanced our understanding of the molecular and cellular bases of haemostasis and thrombosis. However, in vitro assays cannot simultaneously reproduce the interactions of all of the components of the haemostatic process that occur in vivo nor do they reflect the importance of haemodynamic factors resulting from blood flow. To overcome these limitations investigators have increasingly turned to animal models of haemostasis and thrombosis. In this article we describe some advances in the visualisation of platelet and endothelial cell activation and blood coagulation in vivo and review what we have learned from our intravital microscopy experiments using primarily the laser-induced injury model for thrombosis.
Collapse
Affiliation(s)
- L Bellido-Martín
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachussetts, USA
| | | | | | | | | |
Collapse
|
116
|
Evaluation of anti-platelet and anti-thrombotic effects of cilostazol with PFA-100® and Multiplate® whole blood aggregometer in vitro, ex vivo and FeCl3-induced thrombosis models in vivo. Thromb Res 2011; 127:565-70. [PMID: 21420150 DOI: 10.1016/j.thromres.2011.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 02/01/2011] [Accepted: 02/10/2011] [Indexed: 11/22/2022]
Abstract
We evaluate the anti-platelet and anti-thrombotic effects of cilostazol using Multiplate® and PFA-100® in vitro and ex vivo with freshly isolated rat whole blood and in vivo venous and arterial thrombosis models in the same species, in an effort to assess the sensitivity of the whole blood aggregometer assays without potential issues of species differences. In vitro assay of anti-platelet effects of cilostazol against collagen-induced aggregation using Multiplate® produced a graded dose-dependent inhibition curve with IC50 value of 75.4 ± 2.4 μM while it showed a highly sensitive and all-or-none type inhibition response from 25 μM in PFA-100®. Interestingly, cilostazol manifested anti-thrombotic effects in vivo at much lower plasma concentrations than the effective concentrations measured in ex vivo or in vitro aggregation tests using PFA-100® or Multiplate®. In addition, the tail bleeding time measurement demonstrated that rats have lower sensitivity to the anti-platelet effects of cilostazol than mice. These results suggest that the detailed comparative evaluation of whole blood aggregometer assays with anti-thrombotic effects in vivo should be preceded before the application of these methods for the pharmacodynamic studies of anti-thrombotic agents.
Collapse
|
117
|
Abstract
In this issue of Blood, Brill and colleagues demonstrate that von Willebrand factor (VWF) release and subsequent platelet adhesion to endothelial cells are required for thrombus formation in a mouse model of deep venous thrombosis (DVT).
Collapse
|
118
|
Brooks MB, Stokol T, Catalfamo JL. Comparative Hemostasis: Animal Models and New Hemostasis Tests. Clin Lab Med 2011; 31:139-59. [DOI: 10.1016/j.cll.2010.10.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
119
|
Riehl TE, He L, Zheng L, Greco S, Tollefsen DM, Stenson WF. COX-1(+/-)COX-2(-/-) genotype in mice is associated with shortened time to carotid artery occlusion through increased PAI-1. J Thromb Haemost 2011; 9:350-60. [PMID: 21138526 PMCID: PMC3687774 DOI: 10.1111/j.1538-7836.2010.04156.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND We found a high incidence of thrombotic deaths in COX-1(+/-)COX-2(-/-) mice and sought to define the mechanism of these events. The cyclooxygenase products thromboxane A(2) and prostacyclin are important in the regulation of coagulation but their role in fibrinolysis is largely unexplored. PAI-1 blocks fibrinolysis by inhibiting plasminogen activator. AIM Our objective was to explain the mechanism of increased thrombosis associated with the COX-1(+/-)COX-2(-/-) genotype. METHODS Carotid artery occlusion times were measured after photochemical injury. PAI-1 levels were measured in the plasma by ELISA. PAI-1 levels in the aorta were measured by RT-PCR and Western blotting. Urinary metabolites of Thromboxane A(2) and prostacyclin were measured by ELISA. RESULTS The COX-1(+/-)COX-2(-/-) genotype is associated with a decreased time to occlusion in the carotid artery thrombosis model (30 ± 5 minutes vs 60 ± minutes in wild type, p<.001). The COX-1(-/-)COX-2(+/+), COX-1(+/-)COX-2(+/-) and COX-1(+/-)COX-2(+/+) all had occlusion times similar to wild type. COX-1(+/+)COX-2(-/-) had a prolonged occlusion time. COX-1(+/-)COX-2(-/-) had increased PAI-1 levels in the plasma and aorta and with a prolonged euglobulin lysis time (37.4 ± 10.2 hours vs 15.6 ± 9.8 hours in wild type, p<.004). The decreased time to occlusion in the COX-1(+/-)COX2(-/-) mice was normalized by an inhibitory antibody to PAI-1 whereas the antibody had no effect on the time to occlusion in wild type mice. CONCLUSION The COX-1(+/-)COX-2(-/-) genotype is associated with a shortened time to occlusion in the carotid thrombosis model and the shortened time to occlusion is mediated through increased PAI-1 levels resulting in decreased fibrinolysis.
Collapse
Affiliation(s)
- T E Riehl
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
120
|
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is the main inhibitor of tissue-type plasminogen activator (t-PA) and urokinase-type plasminogen activator (u-PA) and therefore plays an important role in the plasminogen/plasmin system. PAI-1 is involved in a variety of cardiovascular diseases (mainly through inhibition of t-PA) as well as in cell migration and tumor development (mainly through inhibition of u-PA and interaction with vitronectin). PAI-1 is a unique member of the serpin superfamily, exhibiting particular unique conformational and functional properties. Since its involvement in various biological and pathophysiological processes PAI-1 has been the subject of many in vivo studies in mouse models. We briefly discuss structural and physiological differences between human and mouse PAI-1 that should be taken into account prior to extrapolation of data obtained in mouse models to the human situation. The current review provides an overview of the various models, with a focus on cardiovascular disease and cancer, using wild-type mice or genetically modified mice, either deficient in PAI-1 or overexpressing different variants of PAI-1.
Collapse
|
121
|
Mitsios JV, Prévost N, Kasirer-Friede A, Gutierrez E, Groisman A, Abrams CS, Wang Y, Litvinov RI, Zemljic-Harpf A, Ross RS, Shattil SJ. What is vinculin needed for in platelets? J Thromb Haemost 2010; 8:2294-304. [PMID: 20670372 PMCID: PMC2965783 DOI: 10.1111/j.1538-7836.2010.03998.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
UNLABELLED Summary. BACKGROUND Vinculin links integrins to the cell cytoskeleton by virtue of its binding to proteins such as talin and F-actin. It has been implicated in the transmission of mechanical forces from the extracellular matrix to the cytoskeleton of migrating cells. Vinculin's function in platelets is unknown. OBJECTIVE To determine whether vinculin is required for the functions of platelets and their major integrin, α(IIb) β(3) . METHODS The murine vinculin gene (Vcl) was deleted in the megakaryocyte/platelet lineage by breeding Vcl fl/fl mice with Pf4-Cre mice. Platelet and integrin functions were studied in vivo and ex vivo. RESULTS Vinculin was undetectable in platelets from Vcl fl/fl Cre(+) mice, as determined by immunoblotting and fluorescence microscopy. Vinculin-deficient megakaryocytes exhibited increased membrane tethers in response to mechanical pulling on α(IIb) β(3) with laser tweezers, suggesting that vinculin helps to maintain membrane cytoskeleton integrity. Surprisingly, vinculin-deficient platelets displayed normal agonist-induced fibrinogen binding to α(IIb) β(3) , aggregation, spreading, actin polymerization/organization, clot retraction and the ability to form a procoagulant surface. Furthermore, vinculin-deficient platelets adhered to immobilized fibrinogen or collagen normally, under both static and flow conditions. Tail bleeding times were prolonged in 59% of vinculin-deficient mice. However, these mice exhibited no spontaneous bleeding and they formed occlusive platelet thrombi comparable to those in wild-type littermates in response to carotid artery injury with FeCl(3) . CONCLUSION Despite promoting membrane cytoskeleton integrity when mechanical force is applied to α(IIb) β(3) , vinculin is not required for the traditional functions of α(IIb) β(3) or the platelet actin cytoskeleton.
Collapse
Affiliation(s)
- John V. Mitsios
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Nicolas Prévost
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Ana Kasirer-Friede
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Edgar Gutierrez
- Department of Physics, University of California, San Diego, La Jolla, CA 92093
| | - Alex Groisman
- Department of Physics, University of California, San Diego, La Jolla, CA 92093
| | - Charles S. Abrams
- Department of Medicine and Cell, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Yanfeng Wang
- Department of Medicine and Cell, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Rustem I. Litvinov
- Department of Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Alice Zemljic-Harpf
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- VA Healthcare San Diego, San Diego, CA 92161
| | - Robert S. Ross
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- VA Healthcare San Diego, San Diego, CA 92161
| | - Sanford J. Shattil
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
122
|
Spontaneous Irs1 passenger mutation linked to a gene-targeted SerpinB2 allele. Proc Natl Acad Sci U S A 2010; 107:16904-9. [PMID: 20837540 DOI: 10.1073/pnas.1012050107] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In characterizing mice with targeted disruption of the SerpinB2 gene, we observed animals that were small at birth with delayed growth and decreased life expectancy. Although this phenotype cosegregated with homozygosity for the inactive SerpinB2 allele, analysis of homozygous SerpinB2-deficient mice derived from two additional independent embryonic stem (ES) cell clones exhibited no growth abnormalities. Examination of additional progeny from the original SerpinB2-deficient line revealed recombination between the small phenotype (smla) and the SerpinB2 locus. The locus responsible for smla was mapped to a 2.78-Mb interval approximately 30 Mb proximal to SerpinB2, bounded by markers D1Mit382 and D1Mit216. Sequencing of Irs1 identified a nonsense mutation at serine 57 (S57X), resulting in complete loss of IRS1 protein expression. Analysis of ES cell DNA suggests that the S57X Irs1 mutation arose spontaneously in an ES cell subclone during cell culture. Although the smla phenotype is similar to previously reported Irs1 alleles, mice exhibited decreased survival, in contrast to the enhanced longevity reported for IRS1 deficiency generated by gene targeting. This discrepancy could result from differences in strain background, unintended indirect effects of the gene targeting, or the minimal genetic interference of the S57X mutation compared with the conventionally targeted Irs1-KO allele. Spontaneous mutations arising during ES cell culture may be a frequent but underappreciated occurrence. When linked to a targeted allele, such mutations could lead to incorrect assignment of phenotype and may account for a subset of markedly discordant results from experiments independently targeting the same gene.
Collapse
|
123
|
Secor D, Li F, Ellis CG, Sharpe MD, Gross PL, Wilson JX, Tyml K. Impaired microvascular perfusion in sepsis requires activated coagulation and P-selectin-mediated platelet adhesion in capillaries. Intensive Care Med 2010; 36:1928-34. [PMID: 20689935 DOI: 10.1007/s00134-010-1969-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Accepted: 05/07/2010] [Indexed: 12/19/2022]
Abstract
PURPOSE Impaired microvascular perfusion in sepsis is not treated effectively because its mechanism is unknown. Since inflammatory and coagulation pathways cross-activate, we tested if stoppage of blood flow in septic capillaries is due to oxidant-dependent adhesion of platelets in these microvessels. METHODS Sepsis was induced in wild type, eNOS(-/-), iNOS(-/-), and gp91phox(-/-) mice (n = 14-199) by injection of feces into the peritoneum. Platelet adhesion, fibrin deposition, and blood flow stoppage in capillaries of hindlimb skeletal muscle were assessed by intravital microscopy. Prophylactic treatments at the onset of sepsis were intravenous injection of platelet-depleting antibody, P-selectin blocking antibody, ascorbate, or antithrombin. Therapeutic treatments (delayed until 6 h) were injection of ascorbate or the glycoprotein IIb/IIIa inhibitor eptifibatide, or local superfusion of the muscle with NOS cofactor tetrahydrobiopterin or NO donor S-nitroso-N-acetylpenicillamine (SNAP). RESULTS Sepsis at 6-7 h markedly increased the number of stopped-flow capillaries and the occurrence of platelet adhesion and fibrin deposition in these capillaries. Platelet depletion, iNOS and gp91phox deficiencies, P-selectin blockade, antithrombin, or prophylactic ascorbate prevented, whereas delayed ascorbate, eptifibatide, tetrahydrobiopterin, or SNAP reversed, septic platelet adhesion and/or flow stoppage. The reversals by ascorbate and tetrahydrobiopterin were absent in eNOS(-/-) mice. Platelet adhesion predicted 90% of capillary flow stoppage. CONCLUSION Impaired perfusion and/or platelet adhesion in septic capillaries requires NADPH oxidase, iNOS, P-selectin, and activated coagulation, and is inhibited by intravenous administration of ascorbate and by local superfusion of tetrahydrobiopterin and NO. Reversal of flow stoppage by ascorbate and tetrahydrobiopterin may depend on local eNOS-derived NO which dislodges platelets from the capillary wall.
Collapse
Affiliation(s)
- Dan Secor
- Critical Illness Research, Victoria Research Laboratories, Lawson Health Research Institute, London, ON, N6C 2V5, Canada
| | | | | | | | | | | | | |
Collapse
|
124
|
Itoh Y, Toriumi H, Yamada S, Hoshino H, Suzuki N. Resident endothelial cells surrounding damaged arterial endothelium reendothelialize the lesion. Arterioscler Thromb Vasc Biol 2010; 30:1725-32. [PMID: 20558819 DOI: 10.1161/atvbaha.110.207365] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To evaluate endothelial repair processes in denuded pial vessels to clarify mechanisms for reconstructing endothelium (because endothelial repair of the cerebral artery after its damage is critical for the prevention of thrombosis, the maintenance of vascular tone, and the protection of the brain by the blood-brain barrier). METHODS AND RESULTS Endothelial cells (ECs) in a 350-microm-long segment of the middle cerebral artery were damaged through a photochemical reaction. Tie2-green fluorescent protein transgenic mice were used for the identification of ECs. Six hours after the endothelial damage, ECs were detached from the luminal surface of the damaged artery, which was then covered with a platelet carpet. Within 24 hours, recovery of the denuded artery started at both edges, with EC elongation and migration. The repair rate was faster at the proximal edge than at the distal edge. Reendothelialization with EC proliferation peaked at 2 to 3 days and ended at 5 days, together with normalization of EC length, with no apparent involvement of foreign progenitor cells. CONCLUSIONS Our in vivo study demonstrated a stepwise reendothelialization process by resident ECs of the pial artery. The prevention of thrombosis, vasospasm, and treatment for blood-brain barrier dysfunction should be considered during the reendothelialization period.
Collapse
Affiliation(s)
- Yoshiaki Itoh
- Department of Neurology, Keio University School of Medicine, Shinanomachi 35, Shinjuku, Tokyo 160-8582, Japan.
| | | | | | | | | |
Collapse
|
125
|
Cortes-Canteli M, Paul J, Norris EH, Bronstein R, Ahn HJ, Zamolodchikov D, Bhuvanendran S, Fenz KM, Strickland S. Fibrinogen and beta-amyloid association alters thrombosis and fibrinolysis: a possible contributing factor to Alzheimer's disease. Neuron 2010; 66:695-709. [PMID: 20547128 PMCID: PMC2895773 DOI: 10.1016/j.neuron.2010.05.014] [Citation(s) in RCA: 281] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2010] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder in which vascular pathology plays an important role. Since the beta-amyloid peptide (Abeta) is a critical factor in this disease, we examined its relationship to fibrin clot formation in AD. In vitro and in vivo experiments showed that fibrin clots formed in the presence of Abeta are structurally abnormal and resistant to degradation. Fibrin(ogen) was observed in blood vessels positive for amyloid in mouse and human AD samples, and intravital brain imaging of clot formation and dissolution revealed abnormal thrombosis and fibrinolysis in AD mice. Moreover, depletion of fibrinogen lessened cerebral amyloid angiopathy pathology and reduced cognitive impairment in AD mice. These experiments suggest that one important contribution of Abeta to AD is via its effects on fibrin clots, implicating fibrin(ogen) as a potential critical factor in this disease.
Collapse
Affiliation(s)
- Marta Cortes-Canteli
- Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Justin Paul
- Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Erin H. Norris
- Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Robert Bronstein
- Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Hyung Jin Ahn
- Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Daria Zamolodchikov
- Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | | | - Katherine M. Fenz
- Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Sidney Strickland
- Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
126
|
Zheng H, Lapointe J, Hekimi S. Lifelong protection from global cerebral ischemia and reperfusion in long-lived Mclk1(+/)(-) mutants. Exp Neurol 2010; 223:557-65. [PMID: 20170652 PMCID: PMC4053415 DOI: 10.1016/j.expneurol.2010.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Revised: 02/01/2010] [Accepted: 02/09/2010] [Indexed: 01/07/2023]
Abstract
To achieve a long life span, animals must be resistant to various injuries as well as avoid or delay lethality from age-dependent diseases. Reduced expression of the mitochondrial enzyme CLK-1/MCLK1 (a.k.a. Coq7), a mitochondrial hydroxylase that is necessary for the biosynthesis of ubiquinone (UQ), extends lifespan in Caenorhabditiselegans and in mice. Here, we show that long-lived Mclk1(+/)(-) mutants have enhanced resistance to neurological damage following global cerebral ischemia-reperfusion (I/R) injury induced by transient bilateral common carotid artery occlusion (BCCAO). Both young ( approximately 100days old) and relatively aged ( approximately 450days old) mutants display increased resistance as indicated by a significant decrease in the amount of degenerating cells observed in forebrain cortex and in hippocampal areas after ischemia and reperfusion. Furthermore, less oxidative damage resulting from the procedure was measured in the brain of young Mclk1(+/)(-) animals. The finding that both young and old mutants are protected indicates that this is a basic phenotype of these mutants and not a secondary consequence of their slow rate of aging. Thus, the partial resistance to I/R injury suggests that Mclk1(+/)(-) mutants have an enhanced recovery potential following age-dependant vascular accidents, which correlates well with their longer survival. By relating this neuroprotective effect to previously reported characteristics of the Mclk1(+/)(-) phenotype, including altered mitochondrial metabolism and increased HIF-1alpha expression, this study establishes these mutants as useful models to analyze the mechanisms underlying tolerance to ischemia, particularly those associated with ischemic preconditioning, as well as to clarify the relation between aging and age-dependent diseases.
Collapse
Affiliation(s)
- Huaien Zheng
- Department of Biology, McGill University, 1205 Avenue Docteur Penfield, Montréal, H3A 1B1, Canada
| | - Jérôme Lapointe
- Department of Biology, McGill University, 1205 Avenue Docteur Penfield, Montréal, H3A 1B1, Canada
| | - Siegfried Hekimi
- Department of Biology, McGill University, 1205 Avenue Docteur Penfield, Montréal, H3A 1B1, Canada
| |
Collapse
|
127
|
Baumgartner B, Jaki T, Wolfsegger MJ, Eder B, Schiviz A, Schwarz HP, Muchitsch EM. Optimization, refinement and reduction of murine in vivo experiments to assess therapeutic approaches for haemophilia A. Lab Anim 2010; 44:211-7. [PMID: 20507877 DOI: 10.1258/la.2010.009113] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The tail cut bleeding model (CUT) is routinely used in factor VIII-deficient mice to assess pharmacodynamic effects of therapeutic strategies for haemophilia A. Results from this model are highly variable, many modifications to the model are reported and at times the animals' wellbeing may be compromised by recording survival as an endpoint. We therefore investigated if the ferric chloride carotid occlusion model (COM) used for thrombosis research can be applied to enhance data quality and animal welfare in haemophilia A research. Relative dose effects and relative dose variations were calculated for the CUT and COM. The requisite sample sizes were estimated and the importance of survival rates to assess rebleeds during recovery was evaluated by correlating initial blood loss to mortality. Relative dose effects increased with higher doses in both models. The COM was more sensitive at lower doses than the CUT, had up to 82% less variation across doses and clearly showed superior accuracy. Only 5% of the sample size required for the CUT would be needed to establish non-inferiority between a specific therapeutic dose in haemophilia A mice and healthy wild-type animals. A strong statistically significant correlation was found between initial blood loss and mortality within 24 h. Our findings clearly suggest that the COM is a valid tool for assessing haemophilia A treatment in vivo. The highly reproducible data means that significantly fewer animals are required and a more humane endpoint can be used by directly assessing clot stability instead of survival rate.
Collapse
Affiliation(s)
- B Baumgartner
- Technology Resources, Baxter Healthcare Corporation, Round Lake, IL, USA.
| | | | | | | | | | | | | |
Collapse
|
128
|
|
129
|
Surin W, Prakash P, Barthwal M, Dikshit M. Optimization of ferric chloride induced thrombosis model in rats: Effect of anti-platelet and anti-coagulant drugs. J Pharmacol Toxicol Methods 2010; 61:287-91. [DOI: 10.1016/j.vascn.2009.11.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Revised: 09/30/2009] [Accepted: 11/23/2009] [Indexed: 11/15/2022]
|
130
|
Maurer LM, Tomasini-Johansson BR, Mosher DF. Emerging roles of fibronectin in thrombosis. Thromb Res 2010; 125:287-91. [PMID: 20116835 PMCID: PMC2877496 DOI: 10.1016/j.thromres.2009.12.017] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 12/01/2009] [Accepted: 12/21/2009] [Indexed: 01/16/2023]
Abstract
Fibronectin (FN) is a glycoprotein recognized originally in the 1940's as a contaminant of fibrinogen in Cohn fraction I of plasma. Decades of research demonstrated FN synthesis by a variety of cells and defined FN as an essential component of the extracellular matrix with roles in embryogenesis, development, and wound healing. More recently, FN has emerged as player in platelet thrombus formation and diseases associated with thrombosis including vascular remodeling, atherosclerosis, and cardiac repair following a myocardial infarct. We discuss the mechanisms by which this might occur and conclude that FN may have a unique role in thrombosis without affecting normal hemostasis and therefore may be a reasonable therapeutic target for the prevention of thrombotic diseases.
Collapse
|
131
|
Freudenberger T, Oppermann M, Marzoll A, Heim HK, Mayer P, Kojda G, Weber AA, Schrör K, Fischer JW. Differential effects of medroxyprogesterone acetate on thrombosis and atherosclerosis in mice. Br J Pharmacol 2010; 158:1951-60. [PMID: 20050187 DOI: 10.1111/j.1476-5381.2009.00470.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE The risk for cardiovascular events including venous and arterial disease and stroke is elevated after treatment with estrogen and medroxyprogesterone acetate (MPA) in postmenopausal women. Here, we have investigated the effect of MPA on arterial thrombosis and atherosclerosis in a murine model of atherosclerosis. EXPERIMENTAL APPROACH Apolipoprotein E (ApoE)-/- mice were bilaterally ovariectomized and treated with placebo, MPA (27.7 microg day(-1)) and MPA + 17-beta-oestradiol (E2; 1.1 microg day(-1)) for 90 days, on a Western-type diet. Thrombotic response was measured in a photothrombosis model, platelet activation by fluorescence activated cell sorting (FACS) analysis (CD62P) and thrombin generation by the endogenous thrombin potential (ETP). Furthermore, aortic plaque burden and aortic root plaque composition were determined. KEY RESULTS MPA and MPA + E2-treated animals showed an aggravated thrombotic response shown by significantly reduced time to stable occlusion. The pro-thrombotic effect of MPA was paralleled by increased ETP whereas platelet activation was not affected. Furthermore, MPA + E2 reduced the number of cells positive for alpha-smooth muscle actin and increased hyaluronan in the plaque matrix. Interestingly, total plaque burden was reduced by MPA but unchanged by MPA + E2. CONCLUSION AND IMPLICATIONS Long-term treatment with MPA and MPA + E2 increased arterial thrombosis despite inhibitory effects of MPA on atherosclerosis in ApoE-deficient mice. Increased thrombin formation, reduced smooth muscle content and remodelling of non-collagenous plaque matrix may be involved in the pro-thrombotic effects. Thus, MPA exhibits differential effects on arterial thrombosis and on atherosclerosis.
Collapse
Affiliation(s)
- Till Freudenberger
- Institut für Pharmakologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Kamocka MM, Mu J, Liu X, Chen N, Zollman A, Sturonas-Brown B, Dunn K, Xu Z, Chen DZ, Alber MS, Rosen ED. Two-photon intravital imaging of thrombus development. JOURNAL OF BIOMEDICAL OPTICS 2010; 15:016020. [PMID: 20210466 PMCID: PMC2844130 DOI: 10.1117/1.3322676] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2008] [Revised: 11/02/2009] [Accepted: 12/18/2009] [Indexed: 05/24/2023]
Abstract
Thrombus development in mouse mesenteric vessels following laser-induced injury was monitored by high-resolution, near-real-time, two-photon, intravital microscopy. In addition to the use of fluorescently tagged fibrin(ogen) and platelets, plasma was labeled with fluorescently tagged dextran. Because blood cells exclude the dextran in the single plane, blood cells appear as black silhouettes. Thus, in addition to monitoring the accumulation of platelets and fibrin in the thrombus, the protocol detects the movement and incorporation of unlabeled cells in and around it. The developing thrombus perturbs the blood flow near the thrombus surface, which affects the incorporation of platelets and blood cells into the structure. The hemodynamic effects and incorporation of blood cells lead to the development of thrombi with heterogeneous domain structures. Additionally, image processing algorithms and simulations were used to quantify structural features of developing thrombi. This analysis suggests a novel mechanism to stop the growth of developing thrombus.
Collapse
Affiliation(s)
- Malgorzata M Kamocka
- Indiana University School of Medicine, Department of Medical and Molecular Genetics, Indianapolis, Indiana 46202, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
White TA, Pan S, Witt TA, Simari RD. Murine strain differences in hemostasis and thrombosis and tissue factor pathway inhibitor. Thromb Res 2010; 125:84-9. [PMID: 19398123 PMCID: PMC2826594 DOI: 10.1016/j.thromres.2009.03.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Revised: 03/18/2009] [Accepted: 03/27/2009] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Differences among murine strains often lead to differential responses in models of human disease. The aim of the current study was to investigate whether differences exist among strains in models of hemostasis and thrombosis and whether these differences are reflected in differences in the tissue factor (TF) pathway. METHODS We examined baseline hemostatic parameters and the response to FeCl3-induced arterial thrombosis and a tail vein bleeding model in C57BL/6J (C57), 129S1/SvImJ (129S), and Balb/cJ (BalbC) mice. Finally, we examined TF and tissue factor pathway inhibitor (TFPI) activities in blood and expression in vascular tissue to determine whether these factors covary with a thrombotic phenotype. RESULTS No differences were observed in PT or aPTT among strains. 129S mice had lower platelet counts (p<0.001). BalbC had an increased rate of occlusion (mean occlusion time of 330+/-45 sec) in a FeCl(3)-induced model of thrombosis when compared to C57 (1182+/-349 sec) or 129 S (1442+/-281 sec) (p<0.05). Similarly, BalbC demonstrated reduced blood loss in tail bleeding experiments when compared to C57 and 129S. Vascular expression of TF and TFPI content did not correlate with the thrombotic phenotype of BalbC. However, circulating TFPI activities were lower in BalbC compared to both C57 and 129S mice. When normalized to circulating TF activities, BalbC had lower circulating TFPI activity than C57 and 129S, and there was a significant correlation between tail bleeding and normalized TFPI activity (r=0.67). CONCLUSIONS These data suggest that there are significant differences among strains in thrombosis and hemostasis and that circulating TFPI activity correlates with these differences.
Collapse
Affiliation(s)
- Thomas A White
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
134
|
Hechler B, Nonne C, Eckly A, Magnenat S, Rinckel JY, Denis CV, Freund M, Cazenave JP, Lanza F, Gachet C. Arterial thrombosis: relevance of a model with two levels of severity assessed by histologic, ultrastructural and functional characterization. J Thromb Haemost 2010; 8:173-84. [PMID: 19874458 DOI: 10.1111/j.1538-7836.2009.03666.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND We previously described a model of laser-induced thrombosis in mesenteric arterioles with superficial and deep levels of injury producing a transient thrombus resolving within 2 min and a larger almost occlusive thrombus, respectively. Both types of lesion were sensitive to platelet GPIIb-IIIa and P2Y(12) inhibition, whereas only deep injuries were sensitive to thrombin blockade. OBJECTIVE The aim of the present study was to use histologic methods and electron and intravital microscopy to characterize the lesions and thrombi and to extend our knowledge of the sensitivity of this model to genetic and pharmacologic inhibition. RESULTS A superficial injury was found to detach the endothelial cells and expose a collagen III- and IV-rich subendothelium where platelets could adhere. Tissue factor and fibrin were not detected. Deeper penetration of the external elastic lamina occurred in deep injuries, with exposure of collagen I, III and IV. Here the thrombus was composed of platelets exhibiting a decreasing gradient of degranulation from the deepest lesion area to the surface. Fibrin was found close to the most activated platelets. Consistently, glycoprotein VI (GPVI)-collagen and GPIb-von Willebrand factor (VWF) interactions were found to be critical in superficial injuries. After deep lesion, thrombus formation was modestly reduced in GPVI-immunodepleted mice and still strongly inhibited in VWF(-/-) mice. Combined hirudin infusion and GPVI depletion further inhibited thrombosis after deep injury. CONCLUSIONS This study confirms the feasibility of inducing arterial thrombosis with distinct levels of severity and establishes the central roles of collagen and VWF in thrombus formation after superficial injury. Collagen, VWF and thrombin all appear to contribute to thrombosis after deep arterial lesion.
Collapse
Affiliation(s)
- B Hechler
- UMR S949 INSERM, Université de Strasbourg, Etablissement Français du Sang-Alsace, Strasbourg, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Abstract
PURPOSE OF REVIEW The thrombotic response to vascular injury is an important clinical problem that mediates most vascular disease complications. Thrombus formation involves an integrated response that is influenced by blood flow, multiple cell types, and numerous circulating factors. As a result, modeling of this complex response using in-vitro or in-silico strategies is insufficient. The use of animal models of thrombosis provides a critical tool for the discovery and initial testing of novel therapies for vascular thrombosis. RECENT FINDINGS The literature from 2008 to the present provides significant advances in regard to novel models of arterial thrombosis, novel mechanisms underlying thrombus formation, new models and mechanisms related to thrombotic stroke, and preclinical advances in therapeutics for vascular thrombosis. SUMMARY The formation of occlusive thrombi is complex, involving the integration of many molecular interactions and cell types at the site of vascular injury. The identification of strategies to suppress occlusive thrombus formation without undermining normal hemostatic function is the primary goal of this area of study.
Collapse
|
136
|
Decrem Y, Rath G, Blasioli V, Cauchie P, Robert S, Beaufays J, Frère JM, Feron O, Dogné JM, Dessy C, Vanhamme L, Godfroid E. Ir-CPI, a coagulation contact phase inhibitor from the tick Ixodes ricinus, inhibits thrombus formation without impairing hemostasis. ACTA ACUST UNITED AC 2009; 206:2381-95. [PMID: 19808248 PMCID: PMC2768864 DOI: 10.1084/jem.20091007] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Blood coagulation starts immediately after damage to the vascular endothelium. This system is essential for minimizing blood loss from an injured blood vessel but also contributes to vascular thrombosis. Although it has long been thought that the intrinsic coagulation pathway is not important for clotting in vivo, recent data obtained with genetically altered mice indicate that contact phase proteins seem to be essential for thrombus formation. We show that recombinant Ixodes ricinus contact phase inhibitor (Ir-CPI), a Kunitz-type protein expressed by the salivary glands of the tick Ixodes ricinus, specifically interacts with activated human contact phase factors (FXIIa, FXIa, and kallikrein) and prolongs the activated partial thromboplastin time (aPTT) in vitro. The effects of Ir-CPI were also examined in vivo using both venous and arterial thrombosis models. Intravenous administration of Ir-CPI in rats and mice caused a dose-dependent reduction in venous thrombus formation and revealed a defect in the formation of arterial occlusive thrombi. Moreover, mice injected with Ir-CPI are protected against collagen- and epinephrine-induced thromboembolism. Remarkably, the effective antithrombotic dose of Ir-CPI did not promote bleeding or impair blood coagulation parameters. To conclude, our results show that a contact phase inhibitor is an effective and safe antithrombotic agent in vivo.
Collapse
Affiliation(s)
- Yves Decrem
- Service de Biologie Moléculaire des Ectoparasites, Institut de Biologie et Médecine Moléculaires, Université Libre de Bruxelles, Gosselies B-6041, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Numaguchi Y, Ishii M, Kubota R, Morita Y, Yamamoto K, Matsushita T, Okumura K, Murohara T. Ablation of angiotensin IV receptor attenuates hypofibrinolysis via PAI-1 downregulation and reduces occlusive arterial thrombosis. Arterioscler Thromb Vasc Biol 2009; 29:2102-8. [PMID: 19745198 DOI: 10.1161/atvbaha.109.195057] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Reduced fibrinolytic activity is associated with adverse cardiovascular events. Although insulin-regulated aminopeptidase (IRAP) was recently identified as the angiotensin (Ang) IV receptor (AT4R), the impact of AngIV-AT4R signaling distal to AngII on the activation of type-1 plasminogen activator inhibitor (PAI-1) in the fibrinolytic process and subsequent formation of thrombosis remains unclarified. METHODS AND RESULTS To determine whether AngIV would inhibit fibrinolysis via PAI-1 activation and promote thrombosis, we evaluated the degree of fibrinolysis in thrombosis models and investigated the roles of AT4R after vascular injury using IRAP knockout mice (IRAP(-/-)). In endothelial cells from control mice (WT; C57Bl6/J), both AngII and AngIV treatments increased PAI-1 mRNA expression in a dose-dependent manner, whereas the response was blunted in endothelial cells from IRAP(-/-) mice. FeCl(3)-induced thrombosis was suppressed in the carotid arteries of IRAP(-/-) mice when compared with WT mice. Similarly, in a model of carotid artery ligation and cuff placement, IRAP(-/-) mice demonstrated accelerated fibrinolysis 7 days after surgery and reduced occlusive thrombosis with negative remodeling at 28 days. CONCLUSIONS AngIV-AT4R signaling has a key role in fibrinolysis and the subsequent formation of arterial thrombosis after vascular injury. AT4R may be a novel therapeutic target against cardiovascular disease.
Collapse
Affiliation(s)
- Yasushi Numaguchi
- Department of Medical Science of Proteases, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
138
|
Abstract
Most inherited hemostatic disorders exhibit incomplete penetrance and variable expressivity, which can be because of genetic or environmental interactions. This wide phenotypic variability for a given disease can be partly explained by modifier gene interactions. Modifier gene interactions have been described for VWD, TTP and venous thrombosis associated with the factor V Leiden mutation. We have exploited advances in mouse genetics in an effort to identify novel genetic loci that may serve as candidate genetic modifiers for bleeding and thrombosis in humans. We have identified several loci affecting plasma VWF levels and have identified and characterized mouse models of ADAMTS13 deficiency and Factor V Leiden that could be useful for identifying novel genes contributing to thrombosis risk in humans.
Collapse
Affiliation(s)
- R J Westrick
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
139
|
Pamuklar Z, Federico L, Liu S, Umezu-Goto M, Dong A, Panchatcharam M, Fulerson Z, Berdyshev E, Natarajan V, Fang X, van Meeteren LA, Moolenaar WH, Mills GB, Morris AJ, Smyth SS, Smyth SS. Autotaxin/lysopholipase D and lysophosphatidic acid regulate murine hemostasis and thrombosis. J Biol Chem 2009; 284:7385-94. [PMID: 19139100 PMCID: PMC2652269 DOI: 10.1074/jbc.m807820200] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The lipid mediator lysophosphatidic acid (LPA) is a potent regulator of vascular cell function in vitro, but its physiologic role in the cardiovasculature is largely unexplored. To address the role of LPA in regulating platelet function and thrombosis, we investigated the effects of LPA on isolated murine platelets. Although LPA activates platelets from the majority of human donors, we found that treatment of isolated murine platelets with physiologic concentrations of LPA attenuated agonist-induced aggregation. Transgenic overexpression of autotaxin/lysophospholipase D (Enpp2), the enzyme necessary for production of the bulk of biologically active LPA in plasma, elevated circulating LPA levels and induced a bleeding diathesis and attenuation of thrombosis in mice. Intravascular administration of exogenous LPA recapitulated the prolonged bleeding time observed in Enpp2-Tg mice. Enpp2+/- mice, which have approximately 50% normal plasma LPA levels, were more prone to thrombosis. Plasma autotaxin associated with platelets during aggregation and concentrated in arterial thrombus, and activated but not resting platelets bound recombinant autotaxin/lysoPLD in an integrin-dependent manner. These results identify a novel pathway in which LPA production by autotaxin/lysoPLD regulates murine hemostasis and thrombosis and suggest that binding of autotaxin/lysoPLD to activated platelets may provide a mechanism to localize LPA production.
Collapse
Affiliation(s)
- Zehra Pamuklar
- Division of Cardiovascular
Medicine, The Gill Heart Institute, and Departments of
Pharmacology and
Biochemistry, University of
Kentucky, Lexington, Kentucky 40536-0200,
Department of Systems Biology, The
University of Texas MD Anderson Cancer Center, Houston, Texas 77030,
Department of Medicine, The University
of Chicago, Chicago, Illinois, 60637,
Department of Biochemistry, Virginia
Commonwealth University School of Medicine, Richmond, Virginia 23298,
Division of Cellular
Biochemistry, Centre for Biomedical Genetics, The Netherlands Cancer
Institute, Plesmanlaan 121, Amsterdam, The Netherlands, and
Department of Veterans Affairs
Medical Center, Lexington, Kentucky 40511
| | - Lorenzo Federico
- Division of Cardiovascular
Medicine, The Gill Heart Institute, and Departments of
Pharmacology and
Biochemistry, University of
Kentucky, Lexington, Kentucky 40536-0200,
Department of Systems Biology, The
University of Texas MD Anderson Cancer Center, Houston, Texas 77030,
Department of Medicine, The University
of Chicago, Chicago, Illinois, 60637,
Department of Biochemistry, Virginia
Commonwealth University School of Medicine, Richmond, Virginia 23298,
Division of Cellular
Biochemistry, Centre for Biomedical Genetics, The Netherlands Cancer
Institute, Plesmanlaan 121, Amsterdam, The Netherlands, and
Department of Veterans Affairs
Medical Center, Lexington, Kentucky 40511
| | - Shuying Liu
- Division of Cardiovascular
Medicine, The Gill Heart Institute, and Departments of
Pharmacology and
Biochemistry, University of
Kentucky, Lexington, Kentucky 40536-0200,
Department of Systems Biology, The
University of Texas MD Anderson Cancer Center, Houston, Texas 77030,
Department of Medicine, The University
of Chicago, Chicago, Illinois, 60637,
Department of Biochemistry, Virginia
Commonwealth University School of Medicine, Richmond, Virginia 23298,
Division of Cellular
Biochemistry, Centre for Biomedical Genetics, The Netherlands Cancer
Institute, Plesmanlaan 121, Amsterdam, The Netherlands, and
Department of Veterans Affairs
Medical Center, Lexington, Kentucky 40511
| | - Makiko Umezu-Goto
- Division of Cardiovascular
Medicine, The Gill Heart Institute, and Departments of
Pharmacology and
Biochemistry, University of
Kentucky, Lexington, Kentucky 40536-0200,
Department of Systems Biology, The
University of Texas MD Anderson Cancer Center, Houston, Texas 77030,
Department of Medicine, The University
of Chicago, Chicago, Illinois, 60637,
Department of Biochemistry, Virginia
Commonwealth University School of Medicine, Richmond, Virginia 23298,
Division of Cellular
Biochemistry, Centre for Biomedical Genetics, The Netherlands Cancer
Institute, Plesmanlaan 121, Amsterdam, The Netherlands, and
Department of Veterans Affairs
Medical Center, Lexington, Kentucky 40511
| | - Anping Dong
- Division of Cardiovascular
Medicine, The Gill Heart Institute, and Departments of
Pharmacology and
Biochemistry, University of
Kentucky, Lexington, Kentucky 40536-0200,
Department of Systems Biology, The
University of Texas MD Anderson Cancer Center, Houston, Texas 77030,
Department of Medicine, The University
of Chicago, Chicago, Illinois, 60637,
Department of Biochemistry, Virginia
Commonwealth University School of Medicine, Richmond, Virginia 23298,
Division of Cellular
Biochemistry, Centre for Biomedical Genetics, The Netherlands Cancer
Institute, Plesmanlaan 121, Amsterdam, The Netherlands, and
Department of Veterans Affairs
Medical Center, Lexington, Kentucky 40511
| | - Manikandan Panchatcharam
- Division of Cardiovascular
Medicine, The Gill Heart Institute, and Departments of
Pharmacology and
Biochemistry, University of
Kentucky, Lexington, Kentucky 40536-0200,
Department of Systems Biology, The
University of Texas MD Anderson Cancer Center, Houston, Texas 77030,
Department of Medicine, The University
of Chicago, Chicago, Illinois, 60637,
Department of Biochemistry, Virginia
Commonwealth University School of Medicine, Richmond, Virginia 23298,
Division of Cellular
Biochemistry, Centre for Biomedical Genetics, The Netherlands Cancer
Institute, Plesmanlaan 121, Amsterdam, The Netherlands, and
Department of Veterans Affairs
Medical Center, Lexington, Kentucky 40511
| | - Zachary Fulerson
- Division of Cardiovascular
Medicine, The Gill Heart Institute, and Departments of
Pharmacology and
Biochemistry, University of
Kentucky, Lexington, Kentucky 40536-0200,
Department of Systems Biology, The
University of Texas MD Anderson Cancer Center, Houston, Texas 77030,
Department of Medicine, The University
of Chicago, Chicago, Illinois, 60637,
Department of Biochemistry, Virginia
Commonwealth University School of Medicine, Richmond, Virginia 23298,
Division of Cellular
Biochemistry, Centre for Biomedical Genetics, The Netherlands Cancer
Institute, Plesmanlaan 121, Amsterdam, The Netherlands, and
Department of Veterans Affairs
Medical Center, Lexington, Kentucky 40511
| | - Evgeny Berdyshev
- Division of Cardiovascular
Medicine, The Gill Heart Institute, and Departments of
Pharmacology and
Biochemistry, University of
Kentucky, Lexington, Kentucky 40536-0200,
Department of Systems Biology, The
University of Texas MD Anderson Cancer Center, Houston, Texas 77030,
Department of Medicine, The University
of Chicago, Chicago, Illinois, 60637,
Department of Biochemistry, Virginia
Commonwealth University School of Medicine, Richmond, Virginia 23298,
Division of Cellular
Biochemistry, Centre for Biomedical Genetics, The Netherlands Cancer
Institute, Plesmanlaan 121, Amsterdam, The Netherlands, and
Department of Veterans Affairs
Medical Center, Lexington, Kentucky 40511
| | - Viswanathan Natarajan
- Division of Cardiovascular
Medicine, The Gill Heart Institute, and Departments of
Pharmacology and
Biochemistry, University of
Kentucky, Lexington, Kentucky 40536-0200,
Department of Systems Biology, The
University of Texas MD Anderson Cancer Center, Houston, Texas 77030,
Department of Medicine, The University
of Chicago, Chicago, Illinois, 60637,
Department of Biochemistry, Virginia
Commonwealth University School of Medicine, Richmond, Virginia 23298,
Division of Cellular
Biochemistry, Centre for Biomedical Genetics, The Netherlands Cancer
Institute, Plesmanlaan 121, Amsterdam, The Netherlands, and
Department of Veterans Affairs
Medical Center, Lexington, Kentucky 40511
| | - Xianjun Fang
- Division of Cardiovascular
Medicine, The Gill Heart Institute, and Departments of
Pharmacology and
Biochemistry, University of
Kentucky, Lexington, Kentucky 40536-0200,
Department of Systems Biology, The
University of Texas MD Anderson Cancer Center, Houston, Texas 77030,
Department of Medicine, The University
of Chicago, Chicago, Illinois, 60637,
Department of Biochemistry, Virginia
Commonwealth University School of Medicine, Richmond, Virginia 23298,
Division of Cellular
Biochemistry, Centre for Biomedical Genetics, The Netherlands Cancer
Institute, Plesmanlaan 121, Amsterdam, The Netherlands, and
Department of Veterans Affairs
Medical Center, Lexington, Kentucky 40511
| | - Laurens A. van Meeteren
- Division of Cardiovascular
Medicine, The Gill Heart Institute, and Departments of
Pharmacology and
Biochemistry, University of
Kentucky, Lexington, Kentucky 40536-0200,
Department of Systems Biology, The
University of Texas MD Anderson Cancer Center, Houston, Texas 77030,
Department of Medicine, The University
of Chicago, Chicago, Illinois, 60637,
Department of Biochemistry, Virginia
Commonwealth University School of Medicine, Richmond, Virginia 23298,
Division of Cellular
Biochemistry, Centre for Biomedical Genetics, The Netherlands Cancer
Institute, Plesmanlaan 121, Amsterdam, The Netherlands, and
Department of Veterans Affairs
Medical Center, Lexington, Kentucky 40511
| | - Wouter H. Moolenaar
- Division of Cardiovascular
Medicine, The Gill Heart Institute, and Departments of
Pharmacology and
Biochemistry, University of
Kentucky, Lexington, Kentucky 40536-0200,
Department of Systems Biology, The
University of Texas MD Anderson Cancer Center, Houston, Texas 77030,
Department of Medicine, The University
of Chicago, Chicago, Illinois, 60637,
Department of Biochemistry, Virginia
Commonwealth University School of Medicine, Richmond, Virginia 23298,
Division of Cellular
Biochemistry, Centre for Biomedical Genetics, The Netherlands Cancer
Institute, Plesmanlaan 121, Amsterdam, The Netherlands, and
Department of Veterans Affairs
Medical Center, Lexington, Kentucky 40511
| | - Gordon B. Mills
- Division of Cardiovascular
Medicine, The Gill Heart Institute, and Departments of
Pharmacology and
Biochemistry, University of
Kentucky, Lexington, Kentucky 40536-0200,
Department of Systems Biology, The
University of Texas MD Anderson Cancer Center, Houston, Texas 77030,
Department of Medicine, The University
of Chicago, Chicago, Illinois, 60637,
Department of Biochemistry, Virginia
Commonwealth University School of Medicine, Richmond, Virginia 23298,
Division of Cellular
Biochemistry, Centre for Biomedical Genetics, The Netherlands Cancer
Institute, Plesmanlaan 121, Amsterdam, The Netherlands, and
Department of Veterans Affairs
Medical Center, Lexington, Kentucky 40511
| | - Andrew J. Morris
- Division of Cardiovascular
Medicine, The Gill Heart Institute, and Departments of
Pharmacology and
Biochemistry, University of
Kentucky, Lexington, Kentucky 40536-0200,
Department of Systems Biology, The
University of Texas MD Anderson Cancer Center, Houston, Texas 77030,
Department of Medicine, The University
of Chicago, Chicago, Illinois, 60637,
Department of Biochemistry, Virginia
Commonwealth University School of Medicine, Richmond, Virginia 23298,
Division of Cellular
Biochemistry, Centre for Biomedical Genetics, The Netherlands Cancer
Institute, Plesmanlaan 121, Amsterdam, The Netherlands, and
Department of Veterans Affairs
Medical Center, Lexington, Kentucky 40511
| | - Susan S. Smyth
- Division of Cardiovascular
Medicine, The Gill Heart Institute, and Departments of
Pharmacology and
Biochemistry, University of
Kentucky, Lexington, Kentucky 40536-0200,
Department of Systems Biology, The
University of Texas MD Anderson Cancer Center, Houston, Texas 77030,
Department of Medicine, The University
of Chicago, Chicago, Illinois, 60637,
Department of Biochemistry, Virginia
Commonwealth University School of Medicine, Richmond, Virginia 23298,
Division of Cellular
Biochemistry, Centre for Biomedical Genetics, The Netherlands Cancer
Institute, Plesmanlaan 121, Amsterdam, The Netherlands, and
Department of Veterans Affairs
Medical Center, Lexington, Kentucky 40511
| | | |
Collapse
|
140
|
Kuijpers MJE, Gilio K, Reitsma S, Nergiz-Unal R, Prinzen L, Heeneman S, Lutgens E, van Zandvoort MAMJ, Nieswandt B, Egbrink MGAO, Heemskerk JWM. Complementary roles of platelets and coagulation in thrombus formation on plaques acutely ruptured by targeted ultrasound treatment: a novel intravital model. J Thromb Haemost 2009; 7:152-61. [PMID: 18983512 DOI: 10.1111/j.1538-7836.2008.03186.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Atherothrombosis is a major cause of cardiovascular events. However, animal models to study this process are scarce. OBJECTIVES We describe the first murine model of acute thrombus formation upon plaque rupture to study atherothrombosis by intravital fluorescence microscopy. METHODS Localized rupture of an atherosclerotic plaque in a carotid artery from Apoe(-/-) mice was induced in vivo using ultrasound. Rupture of the plaque and formation of localized thrombi were verified by two-photon laser scanning microscopy (TPLSM) in isolated arteries, and by immunohistochemistry. The thrombotic reaction was quantified by intravital fluorescence microscopy. RESULTS Inspection of the ultrasound-treated plaques by histochemistry and TPLSM demonstrated local damage, collagen exposure, luminal thrombus formation as well as intra-plaque intrusion of erythrocytes and fibrin. Ultrasound treatment of healthy carotid arteries resulted in endothelial damage and limited platelet adhesion. Real-time intravital fluorescence microscopy demonstrated rapid platelet deposition on plaques and formation of a single thrombus that remained subocclusive. The thrombotic process was antagonized by thrombin inhibition, or by blocking of collagen or adenosine diphosphate receptor pathways. Multiple thrombi were formed in 70% of mice lacking CD40L. CONCLUSIONS Targeted rupture of murine plaques results in collagen exposure and non-occlusive thrombus formation. The thrombotic process relies on platelet activation as well as on thrombin generation and coagulation, and is sensitive to established and novel antithrombotic medication. This model provides new possibilities to study atherothrombosis in vivo.
Collapse
Affiliation(s)
- M J E Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Abstract
There are a myriad of options on where and how to perform thrombosis studies in mice. Models have been developed for systemic thrombosis, larger and smaller vessels of both the arterial and venous systems as well as several different microvascular beds. However, there are important differences between the models and investigators need to be careful and thoughtful when they choose which model to use.
Collapse
Affiliation(s)
- Herbert C Whinna
- Department of Pathology and Laboratory Medicine, School of Medicine, The University of North Carolina at Chapel Hill, NC 27599-7035, USA.
| |
Collapse
|
142
|
Abstract
Hyperhomocysteinemia is an established risk factor for arterial as well as venous thromboembolism. Individuals with severe hyperhomocysteinemia caused by inherited genetic defects in homocysteine metabolism have an extremely high incidence of vascular thrombosis unless they are treated aggressively with homocysteine-lowering therapy. The clinical value of homocysteine-lowering therapy in individuals with moderate hyperhomocysteinemia, which is very common in populations at risk for vascular disease, is more controversial. Considerable progress in our understanding of the molecular mechanisms underlying the association between hyperhomocysteinemia and vascular thrombotic events has been provided by the development of a variety of murine models. Because levels of homocysteine are regulated by both the methionine and folate cycles, hyperhomocysteinemia can be induced in mice through both genetic and dietary manipulations. Mice deficient in the cystathionine beta-synthase (CBS) gene have been exploited widely in many studies investigating the vascular pathophysiology of hyperhomocysteinemia. In this article, we review the established murine models, including the CBS-deficient mouse as well as several newer murine models available for the study of hyperhomocysteinemia. We also summarize the major vascular phenotypes observed in these murine models.
Collapse
Affiliation(s)
- Sanjana Dayal
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, USA
| | | |
Collapse
|
143
|
von zur Muhlen C, von Elverfeldt D, Moeller J, Choudhury R, Paul D, Hagemeyer C, Olschewski M, Becker A, Neudorfer I, Bassler N, Schwarz M, Bode C, Peter K. Magnetic Resonance Imaging Contrast Agent Targeted Toward Activated Platelets Allows In Vivo Detection of Thrombosis and Monitoring of Thrombolysis. Circulation 2008; 118:258-67. [DOI: 10.1161/circulationaha.107.753657] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background—
Platelets are the key to thrombus formation and play a role in the development of atherosclerosis. Noninvasive imaging of activated platelets would be of great clinical interest. Here, we evaluate the ability of a magnetic resonance imaging (MRI) contrast agent consisting of microparticles of iron oxide (MPIOs) and a single-chain antibody targeting ligand-induced binding sites (LIBS) on activated glycoprotein IIb/IIIa to image carotid artery thrombi and atherosclerotic plaques.
Methods and Results—
Anti-LIBS antibody or control antibody was conjugated to 1-μm MPIOs (LIBS MPIO/control MPIO). Nonocclusive mural thrombi were induced in mice with 6% ferric chloride. MRI (at 9.4 T) was performed once before and repeatedly in 12-minute-long sequences after LIBS MPIO/control MPIO injection. After 36 minutes, a significant signal void, corresponding to MPIO accumulation, was observed with LIBS MPIOs but not control MPIOs (
P
<0.05). After thrombolysis, in LIBS MPIO-injected mice, the signal void subsided, indicating successful thrombolysis. On histology, the MPIO content of the thrombus, as well as thrombus size, correlated significantly with LIBS MPIO-induced signal void (both
P
<0.01). After ex vivo incubation of symptomatic human carotid plaques, MRI and histology confirmed binding to areas of platelet adhesion/aggregation for LIBS MPIOs but not for control MPIOs.
Conclusions—
LIBS MPIOs allow in vivo MRI of activated platelets with excellent contrast properties and monitoring of thrombolytic therapy. Furthermore, activated platelets were detected on the surface of symptomatic human carotid plaques by ex vivo MRI. This approach represents a novel noninvasive technique allowing the detection and quantification of platelet-containing thrombi.
Collapse
Affiliation(s)
- C. von zur Muhlen
- From the Departments of Cardiology and Angiology (C.v.z.M., J.A.M., I.N., M.S., C.B.), Radiology/Medical Physics (D.v.E., D.P., A.B.), and Medical Biometry and Statistics (M.O.), University of Freiburg, Freiburg, Germany; Department of Cardiovascular Medicine, University of Oxford, Oxford, UK (R.P.C.); and Baker Heart Research Institute, Melbourne, Australia (C.E.H., N.B., K.P.)
| | - D. von Elverfeldt
- From the Departments of Cardiology and Angiology (C.v.z.M., J.A.M., I.N., M.S., C.B.), Radiology/Medical Physics (D.v.E., D.P., A.B.), and Medical Biometry and Statistics (M.O.), University of Freiburg, Freiburg, Germany; Department of Cardiovascular Medicine, University of Oxford, Oxford, UK (R.P.C.); and Baker Heart Research Institute, Melbourne, Australia (C.E.H., N.B., K.P.)
| | - J.A. Moeller
- From the Departments of Cardiology and Angiology (C.v.z.M., J.A.M., I.N., M.S., C.B.), Radiology/Medical Physics (D.v.E., D.P., A.B.), and Medical Biometry and Statistics (M.O.), University of Freiburg, Freiburg, Germany; Department of Cardiovascular Medicine, University of Oxford, Oxford, UK (R.P.C.); and Baker Heart Research Institute, Melbourne, Australia (C.E.H., N.B., K.P.)
| | - R.P. Choudhury
- From the Departments of Cardiology and Angiology (C.v.z.M., J.A.M., I.N., M.S., C.B.), Radiology/Medical Physics (D.v.E., D.P., A.B.), and Medical Biometry and Statistics (M.O.), University of Freiburg, Freiburg, Germany; Department of Cardiovascular Medicine, University of Oxford, Oxford, UK (R.P.C.); and Baker Heart Research Institute, Melbourne, Australia (C.E.H., N.B., K.P.)
| | - D. Paul
- From the Departments of Cardiology and Angiology (C.v.z.M., J.A.M., I.N., M.S., C.B.), Radiology/Medical Physics (D.v.E., D.P., A.B.), and Medical Biometry and Statistics (M.O.), University of Freiburg, Freiburg, Germany; Department of Cardiovascular Medicine, University of Oxford, Oxford, UK (R.P.C.); and Baker Heart Research Institute, Melbourne, Australia (C.E.H., N.B., K.P.)
| | - C.E. Hagemeyer
- From the Departments of Cardiology and Angiology (C.v.z.M., J.A.M., I.N., M.S., C.B.), Radiology/Medical Physics (D.v.E., D.P., A.B.), and Medical Biometry and Statistics (M.O.), University of Freiburg, Freiburg, Germany; Department of Cardiovascular Medicine, University of Oxford, Oxford, UK (R.P.C.); and Baker Heart Research Institute, Melbourne, Australia (C.E.H., N.B., K.P.)
| | - M. Olschewski
- From the Departments of Cardiology and Angiology (C.v.z.M., J.A.M., I.N., M.S., C.B.), Radiology/Medical Physics (D.v.E., D.P., A.B.), and Medical Biometry and Statistics (M.O.), University of Freiburg, Freiburg, Germany; Department of Cardiovascular Medicine, University of Oxford, Oxford, UK (R.P.C.); and Baker Heart Research Institute, Melbourne, Australia (C.E.H., N.B., K.P.)
| | - A. Becker
- From the Departments of Cardiology and Angiology (C.v.z.M., J.A.M., I.N., M.S., C.B.), Radiology/Medical Physics (D.v.E., D.P., A.B.), and Medical Biometry and Statistics (M.O.), University of Freiburg, Freiburg, Germany; Department of Cardiovascular Medicine, University of Oxford, Oxford, UK (R.P.C.); and Baker Heart Research Institute, Melbourne, Australia (C.E.H., N.B., K.P.)
| | - I. Neudorfer
- From the Departments of Cardiology and Angiology (C.v.z.M., J.A.M., I.N., M.S., C.B.), Radiology/Medical Physics (D.v.E., D.P., A.B.), and Medical Biometry and Statistics (M.O.), University of Freiburg, Freiburg, Germany; Department of Cardiovascular Medicine, University of Oxford, Oxford, UK (R.P.C.); and Baker Heart Research Institute, Melbourne, Australia (C.E.H., N.B., K.P.)
| | - N. Bassler
- From the Departments of Cardiology and Angiology (C.v.z.M., J.A.M., I.N., M.S., C.B.), Radiology/Medical Physics (D.v.E., D.P., A.B.), and Medical Biometry and Statistics (M.O.), University of Freiburg, Freiburg, Germany; Department of Cardiovascular Medicine, University of Oxford, Oxford, UK (R.P.C.); and Baker Heart Research Institute, Melbourne, Australia (C.E.H., N.B., K.P.)
| | - M. Schwarz
- From the Departments of Cardiology and Angiology (C.v.z.M., J.A.M., I.N., M.S., C.B.), Radiology/Medical Physics (D.v.E., D.P., A.B.), and Medical Biometry and Statistics (M.O.), University of Freiburg, Freiburg, Germany; Department of Cardiovascular Medicine, University of Oxford, Oxford, UK (R.P.C.); and Baker Heart Research Institute, Melbourne, Australia (C.E.H., N.B., K.P.)
| | - C. Bode
- From the Departments of Cardiology and Angiology (C.v.z.M., J.A.M., I.N., M.S., C.B.), Radiology/Medical Physics (D.v.E., D.P., A.B.), and Medical Biometry and Statistics (M.O.), University of Freiburg, Freiburg, Germany; Department of Cardiovascular Medicine, University of Oxford, Oxford, UK (R.P.C.); and Baker Heart Research Institute, Melbourne, Australia (C.E.H., N.B., K.P.)
| | - K. Peter
- From the Departments of Cardiology and Angiology (C.v.z.M., J.A.M., I.N., M.S., C.B.), Radiology/Medical Physics (D.v.E., D.P., A.B.), and Medical Biometry and Statistics (M.O.), University of Freiburg, Freiburg, Germany; Department of Cardiovascular Medicine, University of Oxford, Oxford, UK (R.P.C.); and Baker Heart Research Institute, Melbourne, Australia (C.E.H., N.B., K.P.)
| |
Collapse
|
144
|
Barrett NE, Holbrook L, Jones S, Kaiser WJ, Moraes LA, Rana R, Sage T, Stanley RG, Tucker KL, Wright B, Gibbins JM. Future innovations in anti-platelet therapies. Br J Pharmacol 2008; 154:918-39. [PMID: 18587441 PMCID: PMC2451055 DOI: 10.1038/bjp.2008.151] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2008] [Revised: 03/31/2008] [Accepted: 03/31/2008] [Indexed: 12/15/2022] Open
Abstract
Platelets have long been recognized to be of central importance in haemostasis, but their participation in pathological conditions such as thrombosis, atherosclerosis and inflammation is now also well established. The platelet has therefore become a key target in therapies to combat cardiovascular disease. Anti-platelet therapies are used widely, but current approaches lack efficacy in a proportion of patients, and are associated with side effects including problem bleeding. In the last decade, substantial progress has been made in understanding the regulation of platelet function, including the characterization of new ligands, platelet-specific receptors and cell signalling pathways. It is anticipated this progress will impact positively on the future innovations towards more effective and safer anti-platelet agents. In this review, the mechanisms of platelet regulation and current anti-platelet therapies are introduced, and strong, and some more speculative, potential candidate target molecules for future anti-platelet drug development are discussed.
Collapse
Affiliation(s)
- N E Barrett
- School of Biological Sciences, University of Reading, Whiteknights, Reading Berkshire, UK
| | - L Holbrook
- School of Biological Sciences, University of Reading, Whiteknights, Reading Berkshire, UK
| | - S Jones
- School of Biological Sciences, University of Reading, Whiteknights, Reading Berkshire, UK
| | - W J Kaiser
- School of Biological Sciences, University of Reading, Whiteknights, Reading Berkshire, UK
| | - L A Moraes
- School of Biological Sciences, University of Reading, Whiteknights, Reading Berkshire, UK
| | - R Rana
- School of Biological Sciences, University of Reading, Whiteknights, Reading Berkshire, UK
| | - T Sage
- School of Biological Sciences, University of Reading, Whiteknights, Reading Berkshire, UK
| | - R G Stanley
- School of Biological Sciences, University of Reading, Whiteknights, Reading Berkshire, UK
| | - K L Tucker
- School of Biological Sciences, University of Reading, Whiteknights, Reading Berkshire, UK
| | - B Wright
- School of Biological Sciences, University of Reading, Whiteknights, Reading Berkshire, UK
| | - J M Gibbins
- School of Biological Sciences, University of Reading, Whiteknights, Reading Berkshire, UK
| |
Collapse
|
145
|
Wang X. Lipopolysaccharide augments venous and arterial thrombosis in the mouse. Thromb Res 2008; 123:355-60. [PMID: 18448153 DOI: 10.1016/j.thromres.2008.03.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 02/11/2008] [Accepted: 03/13/2008] [Indexed: 10/22/2022]
Abstract
BACKGROUND Animal models of diseases are essential for therapeutic target validation, drug discovery and development. Increasing evidence has demonstrated the importance of inflammation in thrombosis. Here, murine models of vena cava thrombosis and carotid arterial thrombosis augmented by lipopolysaccharide (LPS) were established and characterized to study the association between inflammation and thrombosis. MATERIALS AND METHODS Murine (C57BL/6 mice) models of ferric chloride (FeCl(3))-induced carotid arterial and vena cava thrombosis were established. Thrombus formation was measured indirectly by Doppler blood flow (i.e., clot functional interference with blood flow) in the arterial thrombosis model and directly by protein content of the clot in the venous thrombosis model. An optimal concentration of FeCl(3) was defined to induce thrombus formation and used to study the effects of LPS (i.e., a well-known inflammatory stimulus under these conditions). Real-time polymerase chain reaction (PCR) was used to examine the effect of LPS on TNFalpha and IL-1beta mRNA expression in thrombus formation. RESULTS Dose-dependent analysis demonstrated that 2 mg/kg, i.p., LPS provided a maximal prothrombotic effect in 2.5% ferric chloride-induced vena cava thrombosis, with a 60% increase in thrombus size (n=8, p<0.05) compared to vehicle treatment. In contrast, 2 mg/kg LPS had no significant effect on thrombus formation in a more severe, 3.5% FeCl(3)-induced vena cava thrombosis. A similar prothrombotic effect was observed for LPS in 2.5% FeCl(3)-induced carotid arterial thrombosis model. Treatment of 2 mg/kg LPS significantly augmented arterial thrombosis immediately (between 5-30 minutes) following FeCl(3) injury as assessed by change of Doppler blood flow (n=8, p<0.05). Real-time PCR demonstrated significant induction of TNFalpha and IL-1beta mRNA expression in the thrombus formation in the vessels in response to LPS challenge. CONCLUSION These data demonstrate that LPS augments thrombus formation in acute vascular injury and that LPS-augmented thrombosis might be a useful tool to study the relationship between inflammation and thrombosis.
Collapse
Affiliation(s)
- Xinkang Wang
- Discovery Translational Medicine, Wyeth Research, Collegeville, PA 19426, USA.
| |
Collapse
|