51
|
Verdecchia P, Reboldi G, Angeli F, Mazzotta G, Lip GYH, Brueckmann M, Kleine E, Wallentin L, Ezekowitz MD, Yusuf S, Connolly SJ, Di Pasquale G. Dabigatran vs. warfarin in relation to the presence of left ventricular hypertrophy in patients with atrial fibrillation- the Randomized Evaluation of Long-term anticoagulation therapY (RE-LY) study. Europace 2018; 20:253-262. [PMID: 28520924 PMCID: PMC5834147 DOI: 10.1093/europace/eux022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 02/01/2017] [Indexed: 11/12/2022] Open
Abstract
Aim We tested the hypothesis that left ventricular hypertrophy (LVH) interferes with the antithrombotic effects of dabigatran and warfarin in patients with atrial fibrillation (AF). Methods and results This is a post-hoc analysis of the Randomized Evaluation of Long-term anticoagulation therapY (RE-LY) Study. We defined LVH by electrocardiography (ECG) and included patients with AF on the ECG tracing at entry. Hazard ratios (HR) for each dabigatran dose vs. warfarin were calculated in relation to LVH. LVH was present in 2353 (22.7%) out of 10 372 patients. In patients without LVH, the rates of primary outcome were 1.59%/year with warfarin, 1.60% with dabigatran 110 mg (HR vs. warfarin 1.01, 95% confidence interval (CI) 0.75-1.36) and 1.08% with dabigatran 150 mg (HR vs. warfarin 0.68, 95% CI 0.49-0.95). In patients with LVH, the rates of primary outcome were 3.21%/year with warfarin, 1.69% with dabigatran 110 mg (HR vs. warfarin 0.52, 95% CI 0.32-0.84) and 1.55% with 150 mg (HR vs. warfarin 0.48, 95% CI 0.29-0.78). The interaction between LVH status and dabigatran 110 mg vs. warfarin was significant for the primary outcome (P = 0.021) and stroke (P = 0.016). LVH was associated with a higher event rate with warfarin, not with dabigatran. In the warfarin group, the time in therapeutic range was significantly lower in the presence than in the absence of LVH. Conclusions LVH was associated with a lower antithrombotic efficacy of warfarin, but not of dabigatran, in patients with AF. Consequently, the relative benefit of the lower dose of dabigatran compared to warfarin was enhanced in patients with LVH. The higher dose of dabigatran was superior to warfarin regardless of LVH status. Clinical trial registration http:www.clinicaltrials.gov. Unique identifier: NCT00262600.
Collapse
Affiliation(s)
| | | | - Fabio Angeli
- Department of Cardiology and Cardiovascular Pathophysiology, Hospital S.M. della Misericordia, Perugia, Italy
| | | | - Gregory Y H Lip
- University of Birmingham Institute of Cardiovascular Sciences, City Hospital, Birmingham, UK
| | - Martina Brueckmann
- Boehringer Ingelheim GmbH & Co, Ingelheim am Rhein, Germany.,Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Eva Kleine
- Boehringer Ingelheim GmbH & Co, Ingelheim am Rhein, Germany
| | - Lars Wallentin
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Michael D Ezekowitz
- Sidney Kimmel Medical College at Jefferson University, Philadelphia, PA, USA.,Medical College and Lankenau Medical Center, Wynnewood, PA, USA
| | - Salim Yusuf
- McMaster University, Hamilton, Ontario, Canada
| | | | | |
Collapse
|
52
|
Yang PS, Pak HN, Park DH, Yoo J, Kim TH, Uhm JS, Kim YD, Nam HS, Joung B, Lee MH, Heo JH. Non-cardioembolic risk factors in atrial fibrillation-associated ischemic stroke. PLoS One 2018; 13:e0201062. [PMID: 30028885 PMCID: PMC6054400 DOI: 10.1371/journal.pone.0201062] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 07/06/2018] [Indexed: 02/07/2023] Open
Abstract
Introduction Cardioembolic (CE) risks is usually considered as the main mechanism of ischemic stroke in non-valvular atrial fibrillation (NVAF) patients. However, a substantial number of ischemic strokes in NVAF patients are related to non-CE mechanisms. The aim of this study was to investigate the non-CE risk factors in ischemic stroke patients had NVAF. Methods We included 401 patients (65.6% male, 68.6 ± 9.6 years old) who had been hospitalized due to ischemic stroke and had a known or newly diagnosed NVAF. The CE (intracardiac thrombus, dense spontaneous echo contrast, or low left atrial appendage flow velocity) and non-CE (complex aortic plaque, significant carotid stenosis, or intracranial arterial stenosis) risk factors were investigated at the time of the index stroke. Results The number of CE and non-CE risk factors increased with increasing CHA2DS2-VASc scores (p for trends < 0.001). The presence of CE risk factors was independently associated with persistent atrial fibrillation (p < 0.001), body mass index (p = 0.003), heart failure (p = 0.003), and left atrial volume index (p < 0.001). In contrast, the presence of non-CE risk factors was independently associated with age (p < 0.001), hypertension (p = 0.049), diabetes (p = 0.030), and coronary artery calcium score (CACS; p < 0.001). CACS had the added value in predicting non-CE risk factors of ischemic stroke regardless of the CHA2DS2-VASc risk category (p < 0.001). Conclusion Non-CE risk factors in ischemic stroke patients with NVAF are associated with high CHA2DS2-VASc score and CACS. Atherosclerotic non-CE risk factors should be considered as potential mechanisms of stroke even in patients with AF-associated ischemic stroke.
Collapse
Affiliation(s)
- Pil-Sung Yang
- Division of Cardiology, Department of Internal Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Hui-Nam Pak
- Division of Cardiology, Department of Internal Medicine, Yonsei University Health System, Seoul, Republic of Korea
- * E-mail: (HNP); (JHH)
| | - Dong-Hyuk Park
- Division of Cardiology, Department of Internal Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Joonsang Yoo
- Department of Neurology, Yonsei University Health System, Seoul, Republic of Korea
| | - Tae-Hoon Kim
- Division of Cardiology, Department of Internal Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Jae-Sun Uhm
- Division of Cardiology, Department of Internal Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Young Dae Kim
- Department of Neurology, Yonsei University Health System, Seoul, Republic of Korea
| | - Hyo Suk Nam
- Department of Neurology, Yonsei University Health System, Seoul, Republic of Korea
| | - Boyoung Joung
- Division of Cardiology, Department of Internal Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Moon-Hyoung Lee
- Division of Cardiology, Department of Internal Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Ji Hoe Heo
- Department of Neurology, Yonsei University Health System, Seoul, Republic of Korea
- * E-mail: (HNP); (JHH)
| |
Collapse
|
53
|
Jones SM, Mann A, Conrad K, Saum K, Hall DE, McKinney LM, Robbins N, Thompson J, Peairs AD, Camerer E, Rayner KJ, Tranter M, Mackman N, Owens AP. PAR2 (Protease-Activated Receptor 2) Deficiency Attenuates Atherosclerosis in Mice. Arterioscler Thromb Vasc Biol 2018; 38:1271-1282. [PMID: 29599135 PMCID: PMC6324171 DOI: 10.1161/atvbaha.117.310082] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 03/15/2018] [Indexed: 12/25/2022]
Abstract
OBJECTIVE PAR2 (protease-activated receptor 2)-dependent signaling results in augmented inflammation and has been implicated in the pathogenesis of several autoimmune conditions. The objective of this study was to determine the effect of PAR2 deficiency on the development of atherosclerosis. APPROACH AND RESULTS PAR2 mRNA and protein expression is increased in human carotid artery and mouse aortic arch atheroma versus control carotid and aortic arch arteries, respectively. To determine the effect of PAR2 deficiency on atherosclerosis, male and female low-density lipoprotein receptor-deficient (Ldlr-/-) mice (8-12 weeks old) that were Par2+/+ or Par2-/- were fed a fat- and cholesterol-enriched diet for 12 or 24 weeks. PAR2 deficiency attenuated atherosclerosis in the aortic sinus and aortic root after 12 and 24 weeks. PAR2 deficiency did not alter total plasma cholesterol concentrations or lipoprotein distributions. Bone marrow transplantation showed that PAR2 on nonhematopoietic cells contributed to atherosclerosis. PAR2 deficiency significantly attenuated levels of the chemokines Ccl2 and Cxcl1 in the circulation and macrophage content in atherosclerotic lesions. Mechanistic studies using isolated primary vascular smooth muscle cells showed that PAR2 deficiency is associated with reduced Ccl2 and Cxcl1 mRNA expression and protein release into the supernatant resulting in less monocyte migration. CONCLUSIONS Our results indicate that PAR2 deficiency is associated with attenuation of atherosclerosis and may reduce lesion progression by blunting Ccl2- and Cxcl1-induced monocyte infiltration.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Aortic Diseases/prevention & control
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Carotid Artery Diseases/genetics
- Carotid Artery Diseases/metabolism
- Carotid Artery Diseases/pathology
- Cell Movement
- Cells, Cultured
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Chemokine CXCL1/genetics
- Chemokine CXCL1/metabolism
- Disease Models, Animal
- Female
- Genetic Predisposition to Disease
- Humans
- Lipids/blood
- Macrophages/metabolism
- Macrophages/pathology
- Male
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Plaque, Atherosclerotic
- Receptor, PAR-1/deficiency
- Receptor, PAR-1/genetics
- Receptor, PAR-2/deficiency
- Receptor, PAR-2/genetics
- Receptors, G-Protein-Coupled/deficiency
- Receptors, G-Protein-Coupled/genetics
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
Collapse
Affiliation(s)
- Shannon M Jones
- From the Division of Cardiovascular Health and Disease (S.M.J., A.M., K.C., K.S., L.M.M., N.R., M.T., A.P.O.)
| | - Adrien Mann
- From the Division of Cardiovascular Health and Disease (S.M.J., A.M., K.C., K.S., L.M.M., N.R., M.T., A.P.O.)
| | - Kelsey Conrad
- From the Division of Cardiovascular Health and Disease (S.M.J., A.M., K.C., K.S., L.M.M., N.R., M.T., A.P.O.)
- Pathobiology and Molecular Medicine Program (K.C., M.T., A.P.O.)
| | - Keith Saum
- From the Division of Cardiovascular Health and Disease (S.M.J., A.M., K.C., K.S., L.M.M., N.R., M.T., A.P.O.)
- University of Cincinnati Medical Scientist Training Program (K.S.)
| | - David E Hall
- Department of Nutritional Sciences, College of Allied Health (D.E.H., A.D.P.)
- Department of Internal Medicine (D.E.H., A.D.P.), University of Cincinnati College of Medicine, OH
| | - Lisa M McKinney
- From the Division of Cardiovascular Health and Disease (S.M.J., A.M., K.C., K.S., L.M.M., N.R., M.T., A.P.O.)
| | - Nathan Robbins
- From the Division of Cardiovascular Health and Disease (S.M.J., A.M., K.C., K.S., L.M.M., N.R., M.T., A.P.O.)
| | - Joel Thompson
- Division of Endocrinology and Molecular Medicine, Department of Internal Medicine, University of Kentucky, Lexington (J.T.)
| | - Abigail D Peairs
- Department of Nutritional Sciences, College of Allied Health (D.E.H., A.D.P.)
- Department of Internal Medicine (D.E.H., A.D.P.), University of Cincinnati College of Medicine, OH
| | - Eric Camerer
- INSERM U970, Paris Cardiovascular Research Centre, France (E.C.)
| | - Katey J Rayner
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Michael Tranter
- From the Division of Cardiovascular Health and Disease (S.M.J., A.M., K.C., K.S., L.M.M., N.R., M.T., A.P.O.)
- Pathobiology and Molecular Medicine Program (K.C., M.T., A.P.O.)
| | - Nigel Mackman
- Division of Hematology and Oncology, Department of Medicine, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill (N.M.)
| | - A Phillip Owens
- From the Division of Cardiovascular Health and Disease (S.M.J., A.M., K.C., K.S., L.M.M., N.R., M.T., A.P.O.)
- Pathobiology and Molecular Medicine Program (K.C., M.T., A.P.O.)
| |
Collapse
|
54
|
Kremers BMM, Ten Cate H, Spronk HMH. Pleiotropic effects of the hemostatic system. J Thromb Haemost 2018; 16:S1538-7836(22)02208-5. [PMID: 29851288 DOI: 10.1111/jth.14161] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Indexed: 01/19/2023]
Abstract
Atherothrombosis is characterized by the inflammatory process of atherosclerosis combined with a hypercoagulable state leading to superimposed thrombus formation. In atherosclerotic plaques, cell signaling can occur via protease-activated receptors (PARs), four of which have been identified so far (PAR1-PAR4). Proteases that are able to activate PARs can be produced systemically, but also at the sites of lesions, and they include thrombin and activated factor X. After PAR activation, downstream signaling can lead to both proinflammatory effects and a hypercoagulable state. Which specific effect occurs depends on the type of protease and activated PAR, and the site of activation. Hypercoagulable effects are mainly exerted through PAR1 and PAR4, whereas proinflammatory responses are mostly seen after PAR1 and PAR2 activation. PAR signaling pathways contribute to atherothrombosis, suggesting that inhibition of these pathways possibly prevents cardiovascular events based on this pathophysiological mechanism. In this review, we highlight the pathways by which PAR activation leads to proinflammatory responses and a hypercoagulable state. Furthermore, we give an overview of potential pharmacological treatment targets that promote vascular protection.
Collapse
Affiliation(s)
- B M M Kremers
- Departments of Internal Medicine and Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| | - H Ten Cate
- Departments of Internal Medicine and Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| | - H M H Spronk
- Departments of Internal Medicine and Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
55
|
Raghavan S, Singh NK, Mani AM, Rao GN. Protease-activated receptor 1 inhibits cholesterol efflux and promotes atherogenesis via cullin 3-mediated degradation of the ABCA1 transporter. J Biol Chem 2018; 293:10574-10589. [PMID: 29777060 DOI: 10.1074/jbc.ra118.003491] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 05/10/2018] [Indexed: 12/25/2022] Open
Abstract
Although signaling of thrombin via its receptor protease-activated receptor 1 (Par1) is known to occur in atherothrombosis, its link to the actual pathogenesis of this condition is less clear. To better understand the role of thrombin-Par1 signaling in atherosclerosis, here we have studied their effects on cellular cholesterol efflux in mice. We found that by activating Par1 and cullin 3-mediated ubiquitination and degradation of ABC subfamily A member 1 (ABCA1), thrombin inhibits cholesterol efflux in both murine macrophages and smooth muscle cells. Moreover, disruption of the Par1 gene rescued ABCA1 from Western diet-induced ubiquitination and degradation and restored cholesterol efflux in apolipoprotein E-deficient (ApoE-/-) mice. Similarly, the Par1 deletion diminished diet-induced atherosclerotic lesions in the ApoE-/- mice. These observations for the first time indicate a role for thrombin-Par1 signaling in the pathogenesis of diet-induced atherosclerosis. We identify cullin 3 as a cullin-RING ubiquitin E3 ligase that mediates ABCA1 ubiquitination and degradation and thereby inhibits cholesterol efflux. Furthermore, compared with peripheral blood mononuclear cells (PBMCs) from ApoE-/- mice, the PBMCs from ApoE-/-:Par1-/- mice exhibited decreased trafficking to inflamed arteries of Western diet-fed ApoE-/- mice. This finding suggested that besides inhibiting cholesterol efflux, thrombin-Par1 signaling also plays a role in the recruitment of leukocytes during diet-induced atherogenesis. Based on these findings, we conclude that thrombin-Par1 signaling appears to contribute to the pathogenesis of atherosclerosis by impairing cholesterol efflux from cells and by recruiting leukocytes to arteries.
Collapse
Affiliation(s)
- Somasundaram Raghavan
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Nikhlesh K Singh
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Arul M Mani
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Gadiparthi N Rao
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| |
Collapse
|
56
|
Rana R, Huang T, Koukos G, Fletcher EK, Turner SE, Shearer A, Gurbel PA, Rade JJ, Kimmelstiel CD, Bliden KP, Covic L, Kuliopulos A. Noncanonical Matrix Metalloprotease 1-Protease-Activated Receptor 1 Signaling Drives Progression of Atherosclerosis. Arterioscler Thromb Vasc Biol 2018; 38:1368-1380. [PMID: 29622563 DOI: 10.1161/atvbaha.118.310967] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/22/2018] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Protease-activated receptor-1 (PAR1) is classically activated by thrombin and is critical in controlling the balance of hemostasis and thrombosis. More recently, it has been shown that noncanonical activation of PAR1 by matrix metalloprotease-1 (MMP1) contributes to arterial thrombosis. However, the role of PAR1 in long-term development of atherosclerosis is unknown, regardless of the protease agonist. APPROACH AND RESULTS We found that plasma MMP1 was significantly correlated (R=0.33; P=0.0015) with coronary atherosclerotic burden as determined by angiography in 91 patients with coronary artery disease and acute coronary syndrome undergoing cardiac catheterization or percutaneous coronary intervention. A cell-penetrating PAR1 pepducin, PZ-128, currently being tested as an antithrombotic agent in the acute setting in the TRIP-PCI study (Thrombin Receptor Inhibitory Pepducin-Percutaneous Coronary Intervention), caused a significant decrease in total atherosclerotic burden by 58% to 70% (P<0.05) and reduced plaque macrophage content by 54% (P<0.05) in apolipoprotein E-deficient mice. An MMP1 inhibitor gave similar beneficial effects, in contrast to the thrombin inhibitor bivalirudin that gave no improvement on atherosclerosis end points. Mechanistic studies revealed that inflammatory signaling mediated by MMP1-PAR1 plays a critical role in amplifying tumor necrosis factor α signaling in endothelial cells. CONCLUSIONS These data suggest that targeting the MMP1-PAR1 system may be effective in tamping down chronic inflammatory signaling in plaques and halting the progression of atherosclerosis.
Collapse
Affiliation(s)
- Rajashree Rana
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Tianfang Huang
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Georgios Koukos
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Elizabeth K Fletcher
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Susan E Turner
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Andrew Shearer
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Paul A Gurbel
- Inova Center for Thrombosis Research and Translational Medicine, Inova Heart and Vascular Institute, Inova Fairfax Hospital, Falls Church, VA (P.A.G., K.P.B.)
| | - Jeffrey J Rade
- Department of Medicine, Division of Cardiology, University of Massachusetts Memorial Medical Center, University of Massachusetts Medical School, Worcester (J.J.R.)
| | - Carey D Kimmelstiel
- Department of Medicine, Division of Cardiology, Tufts Medical Center, Boston, MA (C.D.K.)
| | - Kevin P Bliden
- Inova Center for Thrombosis Research and Translational Medicine, Inova Heart and Vascular Institute, Inova Fairfax Hospital, Falls Church, VA (P.A.G., K.P.B.)
| | - Lidija Covic
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| | - Athan Kuliopulos
- From the Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (R.R., T.H., G.K., E.K.F., S.E.T., A.S., L.C., A.K.)
| |
Collapse
|
57
|
Mackman N, Spronk HMH, Stouffer GA, Ten Cate H. Dual Anticoagulant and Antiplatelet Therapy for Coronary Artery Disease and Peripheral Artery Disease Patients. Arterioscler Thromb Vasc Biol 2018; 38:726-732. [PMID: 29449336 PMCID: PMC5978740 DOI: 10.1161/atvbaha.117.310048] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 01/31/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Nigel Mackman
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology (N.M.) and Division of Cardiology (G.A.S.), Department of Medicine, McAllister Heart Institute, University of North Carolina at Chapel Hill; and Laboratory for Clinical Thrombosis and Haemostasis, Thrombosis Expert Centre, Department of Internal Medicine (H.M.H.S., H.t.C.) and Department of Biochemistry (H.M.H.S., H.t.C.), Cardiovascular School of Medicine (Cardiovascular Research Institute Maastricht), Maastricht University Medical Centre, the Netherlands.
| | - Henri M H Spronk
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology (N.M.) and Division of Cardiology (G.A.S.), Department of Medicine, McAllister Heart Institute, University of North Carolina at Chapel Hill; and Laboratory for Clinical Thrombosis and Haemostasis, Thrombosis Expert Centre, Department of Internal Medicine (H.M.H.S., H.t.C.) and Department of Biochemistry (H.M.H.S., H.t.C.), Cardiovascular School of Medicine (Cardiovascular Research Institute Maastricht), Maastricht University Medical Centre, the Netherlands
| | - George A Stouffer
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology (N.M.) and Division of Cardiology (G.A.S.), Department of Medicine, McAllister Heart Institute, University of North Carolina at Chapel Hill; and Laboratory for Clinical Thrombosis and Haemostasis, Thrombosis Expert Centre, Department of Internal Medicine (H.M.H.S., H.t.C.) and Department of Biochemistry (H.M.H.S., H.t.C.), Cardiovascular School of Medicine (Cardiovascular Research Institute Maastricht), Maastricht University Medical Centre, the Netherlands
| | - Hugo Ten Cate
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology (N.M.) and Division of Cardiology (G.A.S.), Department of Medicine, McAllister Heart Institute, University of North Carolina at Chapel Hill; and Laboratory for Clinical Thrombosis and Haemostasis, Thrombosis Expert Centre, Department of Internal Medicine (H.M.H.S., H.t.C.) and Department of Biochemistry (H.M.H.S., H.t.C.), Cardiovascular School of Medicine (Cardiovascular Research Institute Maastricht), Maastricht University Medical Centre, the Netherlands
| |
Collapse
|
58
|
Fabjan A, Bajrović FF. Novel Direct Anticoagulants and Atherosclerosis. Curr Vasc Pharmacol 2018; 17:29-34. [PMID: 29412112 DOI: 10.2174/1570161116666180206111217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 09/13/2017] [Accepted: 11/07/2017] [Indexed: 12/31/2022]
Abstract
Coagulation factors can affect cellular processes that include inflammatory signaling by acting on endothelial protease activated receptors, vascular smooth muscle and inflammatory cells beyond the coagulation cascade. This is important in the pathogenesis of atherosclerosis. Accordingly, experimental data points to beneficial effects of coagulation protease inhibitors on the attenuation of atherosclerosis progression in animal models. However, available clinical data do not support the use of anticoagulants as an add-on treatment of atherosclerosis. New clinical studies are needed with a better selection of patients to clarify the role of novel direct anticoagulants in the management of atherosclerosis.
Collapse
Affiliation(s)
- Andrej Fabjan
- Department of Vascular Neurology and Intensive Care, Neurological Clinic, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Institute of Physiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Fajko F Bajrović
- Department of Vascular Neurology and Intensive Care, Neurological Clinic, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
59
|
Spronk HMH, Padro T, Siland JE, Prochaska JH, Winters J, van der Wal AC, Posthuma JJ, Lowe G, d'Alessandro E, Wenzel P, Coenen DM, Reitsma PH, Ruf W, van Gorp RH, Koenen RR, Vajen T, Alshaikh NA, Wolberg AS, Macrae FL, Asquith N, Heemskerk J, Heinzmann A, Moorlag M, Mackman N, van der Meijden P, Meijers JCM, Heestermans M, Renné T, Dólleman S, Chayouâ W, Ariëns RAS, Baaten CC, Nagy M, Kuliopulos A, Posma JJ, Harrison P, Vries MJ, Crijns HJGM, Dudink EAMP, Buller HR, Henskens YMC, Själander A, Zwaveling S, Erküner O, Eikelboom JW, Gulpen A, Peeters FECM, Douxfils J, Olie RH, Baglin T, Leader A, Schotten U, Scaf B, van Beusekom HMM, Mosnier LO, van der Vorm L, Declerck P, Visser M, Dippel DWJ, Strijbis VJ, Pertiwi K, Ten Cate-Hoek AJ, Ten Cate H. Atherothrombosis and Thromboembolism: Position Paper from the Second Maastricht Consensus Conference on Thrombosis. Thromb Haemost 2018; 118:229-250. [PMID: 29378352 DOI: 10.1160/th17-07-0492] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Atherothrombosis is a leading cause of cardiovascular mortality and long-term morbidity. Platelets and coagulation proteases, interacting with circulating cells and in different vascular beds, modify several complex pathologies including atherosclerosis. In the second Maastricht Consensus Conference on Thrombosis, this theme was addressed by diverse scientists from bench to bedside. All presentations were discussed with audience members and the results of these discussions were incorporated in the final document that presents a state-of-the-art reflection of expert opinions and consensus recommendations regarding the following five topics: 1. Risk factors, biomarkers and plaque instability: In atherothrombosis research, more focus on the contribution of specific risk factors like ectopic fat needs to be considered; definitions of atherothrombosis are important distinguishing different phases of disease, including plaque (in)stability; proteomic and metabolomics data are to be added to genetic information. 2. Circulating cells including platelets and atherothrombosis: Mechanisms of leukocyte and macrophage plasticity, migration, and transformation in murine atherosclerosis need to be considered; disease mechanism-based biomarkers need to be identified; experimental systems are needed that incorporate whole-blood flow to understand how red blood cells influence thrombus formation and stability; knowledge on platelet heterogeneity and priming conditions needs to be translated toward the in vivo situation. 3. Coagulation proteases, fibrin(ogen) and thrombus formation: The role of factor (F) XI in thrombosis including the lower margins of this factor related to safe and effective antithrombotic therapy needs to be established; FXI is a key regulator in linking platelets, thrombin generation, and inflammatory mechanisms in a renin-angiotensin dependent manner; however, the impact on thrombin-dependent PAR signaling needs further study; the fundamental mechanisms in FXIII biology and biochemistry and its impact on thrombus biophysical characteristics need to be explored; the interactions of red cells and fibrin formation and its consequences for thrombus formation and lysis need to be addressed. Platelet-fibrin interactions are pivotal determinants of clot formation and stability with potential therapeutic consequences. 4. Preventive and acute treatment of atherothrombosis and arterial embolism; novel ways and tailoring? The role of protease-activated receptor (PAR)-4 vis à vis PAR-1 as target for antithrombotic therapy merits study; ongoing trials on platelet function test-based antiplatelet therapy adjustment support development of practically feasible tests; risk scores for patients with atrial fibrillation need refinement, taking new biomarkers including coagulation into account; risk scores that consider organ system differences in bleeding may have added value; all forms of oral anticoagulant treatment require better organization, including education and emergency access; laboratory testing still needs rapidly available sensitive tests with short turnaround time. 5. Pleiotropy of coagulation proteases, thrombus resolution and ischaemia-reperfusion: Biobanks specifically for thrombus storage and analysis are needed; further studies on novel modified activated protein C-based agents are required including its cytoprotective properties; new avenues for optimizing treatment of patients with ischaemic stroke are needed, also including novel agents that modify fibrinolytic activity (aimed at plasminogen activator inhibitor-1 and thrombin activatable fibrinolysis inhibitor.
Collapse
Affiliation(s)
- H M H Spronk
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - T Padro
- Cardiovascular Research Center (ICCC), Hospital Sant Pau, Barcelona, Spain
| | - J E Siland
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - J H Prochaska
- Center for Cardiology/Center for Thrombosis and Hemostasis/DZHK, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - J Winters
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - A C van der Wal
- Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands
| | - J J Posthuma
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - G Lowe
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland
| | - E d'Alessandro
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.,Department of Pathology, Academic Medical Center (AMC), Amsterdam, The Netherlands
| | - P Wenzel
- Department of Cardiology, Universitätsmedizin Mainz, Mainz, Germany
| | - D M Coenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - P H Reitsma
- Einthoven Laboratory, Leiden University Medical Center, Leiden, The Netherlands
| | - W Ruf
- Center for Cardiology/Center for Thrombosis and Hemostasis/DZHK, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - R H van Gorp
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - R R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - T Vajen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - N A Alshaikh
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - A S Wolberg
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
| | - F L Macrae
- Thrombosis and Tissue Repair Group, Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - N Asquith
- Thrombosis and Tissue Repair Group, Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - J Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - A Heinzmann
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - M Moorlag
- Synapse, Maastricht, The Netherlands
| | - N Mackman
- Department of Medicine, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina, United States
| | - P van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - J C M Meijers
- Department of Plasma Proteins, Sanquin, Amsterdam, The Netherlands
| | - M Heestermans
- Einthoven Laboratory, Leiden University Medical Center, Leiden, The Netherlands
| | - T Renné
- Department of Molecular Medicine and Surgery, Karolinska Institutet and University Hospital, Stockholm, Sweden.,Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - S Dólleman
- Department of Nephrology, Leiden University Medical Centre, Leiden, The Netherlands
| | - W Chayouâ
- Synapse, Maastricht, The Netherlands
| | - R A S Ariëns
- Thrombosis and Tissue Repair Group, Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - C C Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - M Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - A Kuliopulos
- Tufts University School of Graduate Biomedical Sciences, Biochemistry/Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts
| | - J J Posma
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - P Harrison
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - M J Vries
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - H J G M Crijns
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - E A M P Dudink
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - H R Buller
- Department of Vascular Medicine, Academic Medical Center (AMC), Amsterdam, The Netherlands
| | - Y M C Henskens
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - A Själander
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - S Zwaveling
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands.,Synapse, Maastricht, The Netherlands
| | - O Erküner
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - J W Eikelboom
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - A Gulpen
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - F E C M Peeters
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - J Douxfils
- Department of Pharmacy, Thrombosis and Hemostasis Center, Faculty of Medicine, Namur University, Namur, Belgium
| | - R H Olie
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - T Baglin
- Department of Haematology, Addenbrookes Hospital Cambridge, Cambridge, United Kingdom
| | - A Leader
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands.,Davidoff Cancer Center, Rabin Medical Center, Institute of Hematology, Sackler Faculty of Medicine, Tel Aviv University, Petah Tikva, Tel Aviv, Israel
| | - U Schotten
- Center for Cardiology/Center for Thrombosis and Hemostasis/DZHK, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - B Scaf
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - H M M van Beusekom
- Department of Experimental Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - L O Mosnier
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, United States
| | | | - P Declerck
- Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | | | - D W J Dippel
- Department of Neurology, Erasmus MC, Rotterdam, The Netherlands
| | | | - K Pertiwi
- Department of Cardiovascular Pathology, University of Amsterdam, Academic Medical Center, Amsterdam, The Netherlands
| | - A J Ten Cate-Hoek
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - H Ten Cate
- Laboratory for Clinical Thrombosis and Haemostasis, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
60
|
Nakase T, Moroi J, Ishikawa T. Anti-inflammatory and antiplatelet effects of non-vitamin K antagonist oral anticoagulants in acute phase of ischemic stroke patients. Clin Transl Med 2018; 7:2. [PMID: 29335786 PMCID: PMC5768575 DOI: 10.1186/s40169-017-0179-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 12/22/2017] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Recently, non-vitamin K antagonist oral anticoagulants such as direct thrombin and direct factor Xa inhibitors have been prescribed for prevention of embolic stroke. While in Japan, argatroban, also a direct thrombin inhibitor, is available for the treatment of atherothrombotic stroke patients. This study aimed to explore whether there is any differences between direct thrombin and direct factor Xa inhibitors regarding the inhibiting effect against thrombogenesis in the clinical setting of acute ischemic stroke. METHODS Acute ischemic stroke patients newly prescribed anti-thrombotic agents were consecutively screened, and 44 patients with single medicine were enrolled (median 72.0 years-old). Blood samples were obtained at 1 and 2 weeks after the medication started. The extent of anticoagulation activity, inflammatory markers and platelet aggregation were assessed. Patients with antiplatelets were used as control. RESULTS Prescribed antithrombotics were dabigatran (group D: n = 12), apixaban (group A: n = 14) and antiplatelet agents (group P: n = 18). Prevalence of stroke risks and anticoagulation activity were not different between groups D and A. The alteration of inflammatory markers in a week in the group A showed similar trend to those in the group P. The group D presented relatively lower amount of high-sensitive C-reactive protein and higher amount of pentraxin-3 compared with groups A and P. While 88.9% of group P patients showed decreased platelet aggregation activity with adenosine diphosphate, 55.6% of group D and 40.0% of group A presented the inhibition of platelet aggregation activity. CONCLUSIONS Even in acute ischemic stroke patients, both apixaban and dabigatran equally showed the anticoagulation activity. The reduction of inflammatory response might be prominent in apixaban, whereas the inhibition of platelet aggregation activity might be evident in dabigatran.
Collapse
Affiliation(s)
- Taizen Nakase
- Department of Neurology, Research Institute for Brain and Blood Vessels-Akita, 6-10 Sensyu Kubota Machi, Akita, 010-0874, Japan.
| | - Junta Moroi
- Department of Surgical Neurology, Research Institute for Brain and Blood Vessels-Akita, Akita, Japan
| | - Tatsuya Ishikawa
- Department of Surgical Neurology, Research Institute for Brain and Blood Vessels-Akita, Akita, Japan
| |
Collapse
|
61
|
Animal models of atherosclerosis. Eur J Pharmacol 2017; 816:3-13. [DOI: 10.1016/j.ejphar.2017.05.010] [Citation(s) in RCA: 296] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 04/07/2017] [Accepted: 05/04/2017] [Indexed: 12/31/2022]
|
62
|
Esmon CT. Targeting factor Xa and thrombin: impact on coagulation and beyond. Thromb Haemost 2017; 111:625-33. [DOI: 10.1160/th13-09-0730] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 11/07/2013] [Indexed: 12/11/2022]
Abstract
SummaryGreat advances have been made in recent years in understanding the haemostatic system and the molecular and cellular basis of thrombus formation. Although directly targeting factor Xa or thrombin (factor IIa) for effective anticoagulation is now well established, evidence has emerged suggesting that factor Xa and thrombin are involved in other physiological and pathophysiological cellular processes, including inflammation. These non-haemostatic activities of factor Xa and thrombin are predominantly mediated via the activation of proteinaseactivated receptors. Studies have indicated a potential role of coagulation proteins (including factor Xa and thrombin) in the progression of disease conditions such as atherothrombosis. Preclinical studies have provided evidence for the effects of direct factor Xa or direct thrombin inhibition beyond anticoagulation, including anti-inflammatory activities and atherosclerotic plaque stabilisation. In this article, the non-haemostatic activities of factor Xa and thrombin and the effects of direct inhibition of these coagulation factors on these activities are summarised. In addition, the potential roles of factor Xa and thrombin in atherosclerosis and atherothrombosis are explored and the cardiovascular profiles of rivaroxaban, apixaban and dabigatran etexilate observed in phase III clinical studies are discussed.
Collapse
|
63
|
Amara A, Mrad M, Sayeh A, Haggui A, Lahideb D, Fekih-Mrissa N, Haouala H, Nsiri B. Association of FV G1691A Polymorphism but not A4070G With Coronary Artery Disease. Clin Appl Thromb Hemost 2017; 24:330-337. [PMID: 29179580 PMCID: PMC6714679 DOI: 10.1177/1076029617744320] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Coronary artery disease (CAD) is one of the chief causes of death in the world. Several
hypotheses have been promoted as for the origin of the disease, among which are genetic
predispositions and/or environmental factors. The aim of this study was to determine the
effect of factor V (FV) gene polymorphisms (Leiden, G1691A [FVL] and HR2 A4070G) and to
analyze their association with traditional risk factors in assessing the risk of CAD. Our
study population included 200 Tunisian patients with symptomatic CAD and a control group
of 300 participants matched for age and sex. All participants were genotyped for the FVL
and HR2 polymorphisms. Multivariate logistic regression was applied to analyze independent
factors associated with the risk of CAD. Our analysis showed that the FVL A allele
frequency (P < 10–3, odds ratio [OR] = 2.81, 95% confidence
interval [CI] = 1.6-4.9) and GA genotype (P < 10–3, OR =
4.03, 95% CI = 2.1-7.6) are significantly more prevalent among patients with CAD compared
to those controls and may be predisposing to CAD. We further found that the FVL mutation
is an independent risk factor whose effect is not modified by other factors (smoking,
diabetes, hypertension, dyslipidemia, and a family history of CAD) in increasing the risk
of the disease. However, analysis of FV HR2 variation does not show any statistically
significant association with CAD. The FVL polymorphism may be an independent risk factor
for CAD. However, further investigations on these polymorphisms and their possible
synergisms with traditional risk factors for CAD could help to ascertain better
predictability for CAD susceptibility.
Collapse
Affiliation(s)
- Ahmed Amara
- 1 Hôpital Militaire de Tunis, Service d'Hématologie, Laboratoire de Biologie Moléculaire, Montfleury, Tunisie.,2 Université Tunis el Manar, Faculté des Sciences de Tunis, Tunisie
| | - Meriem Mrad
- 1 Hôpital Militaire de Tunis, Service d'Hématologie, Laboratoire de Biologie Moléculaire, Montfleury, Tunisie.,2 Université Tunis el Manar, Faculté des Sciences de Tunis, Tunisie
| | - Aicha Sayeh
- 1 Hôpital Militaire de Tunis, Service d'Hématologie, Laboratoire de Biologie Moléculaire, Montfleury, Tunisie.,2 Université Tunis el Manar, Faculté des Sciences de Tunis, Tunisie
| | - Abdeddayem Haggui
- 3 Hôpital Militaire de Tunis, Service de Cardiologie, Montfleury, Tunisie.,4 Université de Tunis El Manar, Faculté de Médecine de Tunis, Tunisie
| | - Dhaker Lahideb
- 3 Hôpital Militaire de Tunis, Service de Cardiologie, Montfleury, Tunisie.,4 Université de Tunis El Manar, Faculté de Médecine de Tunis, Tunisie
| | - Najiba Fekih-Mrissa
- 1 Hôpital Militaire de Tunis, Service d'Hématologie, Laboratoire de Biologie Moléculaire, Montfleury, Tunisie.,5 Académie Militaire Fondouk Jédid, Nabeul, Tunisie
| | - Habib Haouala
- 3 Hôpital Militaire de Tunis, Service de Cardiologie, Montfleury, Tunisie.,4 Université de Tunis El Manar, Faculté de Médecine de Tunis, Tunisie
| | - Brahim Nsiri
- 1 Hôpital Militaire de Tunis, Service d'Hématologie, Laboratoire de Biologie Moléculaire, Montfleury, Tunisie.,6 Université de Monastir, Faculté de Pharmacie, Monastir, Tunisie
| |
Collapse
|
64
|
Low dose of alcohol attenuates pro-atherosclerotic activity of thrombin. Atherosclerosis 2017; 265:215-224. [DOI: 10.1016/j.atherosclerosis.2017.09.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/29/2017] [Accepted: 09/01/2017] [Indexed: 01/11/2023]
|
65
|
Dong A, Mueller P, Yang F, Yang L, Morris A, Smyth SS. Direct thrombin inhibition with dabigatran attenuates pressure overload-induced cardiac fibrosis and dysfunction in mice. Thromb Res 2017; 159:58-64. [PMID: 28982031 DOI: 10.1016/j.thromres.2017.09.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/29/2017] [Accepted: 09/19/2017] [Indexed: 12/31/2022]
Abstract
INTRODUCTION The multifunctional serine protease thrombin exerts proinflammatory and profibrotic cellular effects that may contribute to cardiac remodeling. This study was designed to investigate whether direct thrombin inhibition with dabigatran attenuates myocardial injury in the setting of pressure overload-induced heart failure. MATERIAL AND METHODS Transverse aortic constriction (TAC) surgery was performed on C57Bl/6J male mice to elicit cardiac hypertrophy. TAC, or sham, mice were randomly assigned to receive chow supplemented with the oral anticoagulant, dabigatran etexilate, or placebo. RESULTS Dabigatran did not affect cardiac hypertrophy, as measured by heart weight-to-body weight or the heart weight-to-tibia length, although a non-significant reduction in myocardial hypertrophic markers (ANP, BNP and MHC) occurred. Dabigatran reduced perivascular fibrosis by 25%, interstitial fibrosis by 54%, and the expression of myocardial fibrosis markers collagen I & III, MMP9, SMA, and PAR-1. These changes were associated with significant improvement in both coronary flow reserve and global left ventricular function. In cultured cardiac fibroblasts, dabigatran decreased thrombin and PAR-1-mediated collagen deposition by 30% and 37%, respectively. CONCLUSIONS Dabigatran attenuates cardiac fibrosis in the setting of pressure overload and improves coronary flow reserve and global cardiac function possibly by inhibiting thrombin activity and down-regulating PAR-1 expression in the absence of an effect on cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Anping Dong
- Division of Cardiovascular Medicine, The Gill Heart Institute, 741 S. Limestone Street, 252 BBSRB, Lexington, KY 40536-0509, United States; Lexington VA Medical Center, 1101 Veterans Drive, Lexington, KY 40502, United States
| | - Paul Mueller
- Division of Cardiovascular Medicine, The Gill Heart Institute, 741 S. Limestone Street, 252 BBSRB, Lexington, KY 40536-0509, United States.
| | - Fanmuyi Yang
- Division of Cardiovascular Medicine, The Gill Heart Institute, 741 S. Limestone Street, 252 BBSRB, Lexington, KY 40536-0509, United States.
| | - Liping Yang
- Division of Cardiovascular Medicine, The Gill Heart Institute, 741 S. Limestone Street, 252 BBSRB, Lexington, KY 40536-0509, United States.
| | - Andrew Morris
- Division of Cardiovascular Medicine, The Gill Heart Institute, 741 S. Limestone Street, 252 BBSRB, Lexington, KY 40536-0509, United States; Lexington VA Medical Center, 1101 Veterans Drive, Lexington, KY 40502, United States.
| | - Susan S Smyth
- Division of Cardiovascular Medicine, The Gill Heart Institute, 741 S. Limestone Street, 252 BBSRB, Lexington, KY 40536-0509, United States; Lexington VA Medical Center, 1101 Veterans Drive, Lexington, KY 40502, United States.
| |
Collapse
|
66
|
Isermann B. Homeostatic effects of coagulation protease-dependent signaling and protease activated receptors. J Thromb Haemost 2017; 15:1273-1284. [PMID: 28671351 DOI: 10.1111/jth.13721] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A homeostatic function of the coagulation system in regard to hemostasis is well established. Homeostasis of blood coagulation depends partially on protease activated receptor (PAR)-signaling. Beyond coagulation proteases, numerous other soluble and cell-bound proteases convey cellular effects via PAR signaling. As we learn more about the mechanisms underlying cell-, tissue-, and context-specific PAR signaling, we concurrently gain new insights into physiological and pathophysiological functions of PARs. In this regard, regulation of cell and tissue homeostasis by PAR signaling is an evolving scheme. Akin to the control of blood clotting per se (the fibrin-platelet interaction) coagulation proteases coordinately regulate cell- and tissue-specific functions. This review summarizes recent insights into homeostatic regulation through PAR signaling, focusing on blood coagulation proteases. Considering the common use of drugs altering coagulation protease activity through either broad or targeted inhibitory activities, and the advent of PAR modulating drugs, an in-depth understanding of the mechanisms through which coagulation proteases and PAR signaling regulate not only hemostasis, but also cell and tissue homeostasis is required.
Collapse
Affiliation(s)
- B Isermann
- Institute of Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| |
Collapse
|
67
|
Ten Cate H, Hackeng TM, García de Frutos P. Coagulation factor and protease pathways in thrombosis and cardiovascular disease. Thromb Haemost 2017; 117:1265-1271. [PMID: 28594052 DOI: 10.1160/th17-02-0079] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/27/2017] [Indexed: 02/06/2023]
Abstract
The biochemical characterisation of the proteolytic pathways that constitute blood coagulation was one of the most relevant achievements in biomedical research during the second half of the 20th century. Understanding these pathways was of crucial importance for improving global health through application in haemostasis and thrombosis pathologies. Immediately after the cloning of the genes corresponding to these proteins, mutations were discovered in them that were associated with imbalances in haemostasis. Later, the importance of coagulation pathways in other pathological processes was demonstrated, such as in atherosclerosis and inflammation, both essential processes involved in vascular disease. In the present review we evaluate the concepts that have allowed us to reach the integrated vision on coagulation that we have today. The thrombo-inflammation model encompassing these aspects includes a pivotal role for the proteases of the coagulation pathway as well as the regulatory proteins thereof. These concepts illustrate the importance of the coagulation cascade in cardiovascular pathology, not only in thrombotic processes, but also in atherosclerotic processes and in the response to ischaemia-reperfusion injury, making it a central mechanism in cardiovascular disease.
Collapse
Affiliation(s)
| | | | - Pablo García de Frutos
- Dr. Pablo García de Frutos, Institute of Biomedical Research of Barcelona (IIBB-CSIC), C/Roselló 161, 08036 Barcelona, Spain, Tel.: +34 933632382, E-mail:
| |
Collapse
|
68
|
Ouweneel AB, Heestermans M, Verwilligen RAF, Gijbels MJJ, Reitsma PH, Van Eck M, van Vlijmen BJM. Silencing of Anticoagulant Protein C Evokes Low-Incident but Spontaneous Atherothrombosis in Apolipoprotein E-Deficient Mice-Brief Report. Arterioscler Thromb Vasc Biol 2017; 37:782-785. [PMID: 28302625 DOI: 10.1161/atvbaha.117.309188] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 03/01/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Murine atherosclerosis models do not spontaneously develop atherothrombotic complications. We investigated whether disruption of natural anticoagulation allows preexisting atherosclerotic plaques to progress toward an atherothrombotic phenotype. APPROACH AND RESULTS On lowering of plasma protein C levels with small interfering RNA (siProc) in 8-week Western-type diet-fed atherosclerotic apolipoprotein E-deficient mice, 1 out of 4 mice displayed a large, organized, and fibrin- and leukocyte-rich thrombus on top of an advanced atherosclerotic plaque located in the aortic root. Although again at low incidence (3 in 25), comparable thrombi at the same location were observed during a second independent experiment in 9-week Western-type diet-fed apolipoprotein E-deficient mice. Mice with thrombi on their atherosclerotic plaques did not show other abnormalities and had equally lowered plasma protein C levels as siProc-treated apolipoprotein E-deficient mice without thrombi. Fibrinogen and thrombin-antithrombin concentrations and blood platelet numbers were also comparable, and plaques in siProc mice with thrombi had a similar composition and size as plaques in siProc mice without thrombi. Seven out of 25 siProc mice featured clots in the left atrium of the heart. CONCLUSIONS Our findings indicate that small interfering RNA-mediated silencing of protein C in apolipoprotein E-deficient mice creates a condition that allows the occurrence of spontaneous atherothrombosis, albeit at a low incidence. Lowering natural anticoagulation in atherosclerosis models may help to discover factors that increase atherothrombotic complications.
Collapse
Affiliation(s)
- Amber B Ouweneel
- From the Division of Biopharmaceutics, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands (A.B.O., R.A.F.V., M.v.E.); Einthoven Laboratory for Experimental Vascular Medicine (M.H., P.H.R., B.J.M.v.V.), and Department of Internal Medicine, Division of Thrombosis and Hemostasis (M.H., P.H.R., B.J.M.v.V.), Leiden University Medical Center, The Netherlands; Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, The Netherlands (M.J.J.G.); and Department of Medical Biochemistry, The Academic Medical Center, Amsterdam, The Netherlands (M.J.J.G.)
| | - Marco Heestermans
- From the Division of Biopharmaceutics, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands (A.B.O., R.A.F.V., M.v.E.); Einthoven Laboratory for Experimental Vascular Medicine (M.H., P.H.R., B.J.M.v.V.), and Department of Internal Medicine, Division of Thrombosis and Hemostasis (M.H., P.H.R., B.J.M.v.V.), Leiden University Medical Center, The Netherlands; Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, The Netherlands (M.J.J.G.); and Department of Medical Biochemistry, The Academic Medical Center, Amsterdam, The Netherlands (M.J.J.G.)
| | - Robin A F Verwilligen
- From the Division of Biopharmaceutics, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands (A.B.O., R.A.F.V., M.v.E.); Einthoven Laboratory for Experimental Vascular Medicine (M.H., P.H.R., B.J.M.v.V.), and Department of Internal Medicine, Division of Thrombosis and Hemostasis (M.H., P.H.R., B.J.M.v.V.), Leiden University Medical Center, The Netherlands; Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, The Netherlands (M.J.J.G.); and Department of Medical Biochemistry, The Academic Medical Center, Amsterdam, The Netherlands (M.J.J.G.)
| | - Marion J J Gijbels
- From the Division of Biopharmaceutics, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands (A.B.O., R.A.F.V., M.v.E.); Einthoven Laboratory for Experimental Vascular Medicine (M.H., P.H.R., B.J.M.v.V.), and Department of Internal Medicine, Division of Thrombosis and Hemostasis (M.H., P.H.R., B.J.M.v.V.), Leiden University Medical Center, The Netherlands; Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, The Netherlands (M.J.J.G.); and Department of Medical Biochemistry, The Academic Medical Center, Amsterdam, The Netherlands (M.J.J.G.)
| | - Pieter H Reitsma
- From the Division of Biopharmaceutics, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands (A.B.O., R.A.F.V., M.v.E.); Einthoven Laboratory for Experimental Vascular Medicine (M.H., P.H.R., B.J.M.v.V.), and Department of Internal Medicine, Division of Thrombosis and Hemostasis (M.H., P.H.R., B.J.M.v.V.), Leiden University Medical Center, The Netherlands; Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, The Netherlands (M.J.J.G.); and Department of Medical Biochemistry, The Academic Medical Center, Amsterdam, The Netherlands (M.J.J.G.)
| | - Miranda Van Eck
- From the Division of Biopharmaceutics, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands (A.B.O., R.A.F.V., M.v.E.); Einthoven Laboratory for Experimental Vascular Medicine (M.H., P.H.R., B.J.M.v.V.), and Department of Internal Medicine, Division of Thrombosis and Hemostasis (M.H., P.H.R., B.J.M.v.V.), Leiden University Medical Center, The Netherlands; Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, The Netherlands (M.J.J.G.); and Department of Medical Biochemistry, The Academic Medical Center, Amsterdam, The Netherlands (M.J.J.G.)
| | - Bart J M van Vlijmen
- From the Division of Biopharmaceutics, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands (A.B.O., R.A.F.V., M.v.E.); Einthoven Laboratory for Experimental Vascular Medicine (M.H., P.H.R., B.J.M.v.V.), and Department of Internal Medicine, Division of Thrombosis and Hemostasis (M.H., P.H.R., B.J.M.v.V.), Leiden University Medical Center, The Netherlands; Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, The Netherlands (M.J.J.G.); and Department of Medical Biochemistry, The Academic Medical Center, Amsterdam, The Netherlands (M.J.J.G.).
| |
Collapse
|
69
|
Parenteral administration of factor Xa/IIa inhibitors limits experimental aortic aneurysm and atherosclerosis. Sci Rep 2017; 7:43079. [PMID: 28220880 PMCID: PMC5318894 DOI: 10.1038/srep43079] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 01/12/2017] [Indexed: 12/20/2022] Open
Abstract
Intraluminal thrombus is a consistent feature of human abdominal aortic aneurysm (AAA). Coagulation factor Xa (FXa) catalyses FII to thrombin (FIIa). We examined the effect of FXa/FIIa inhibition on experimental aortic aneurysm in apolipoprotein E-deficient (ApoE-/-) mice infused with angiotensin II (AngII). The concentration of FXa within the supra-renal aorta (SRA) correlated positively with SRA diameter. Parenteral administration of enoxaparin (FXa/IIa inhibitor) and fondaparinux (FXa inhibitor) over 14 days reduced to severity of aortic aneurysm and atherosclerosis in AngII-infused ApoE-/- mice. Enteral administration of the FIIa inhibitor dabigatran had no significant effect. Aortic protease-activated receptor (PAR)-2 expression increased in response to AngII infusion. Fondaparinux reduced SRA levels of FXa, FIIa, PAR-2, matrix metalloproteinase (MMP)2, Smad2/3 phosphorylation, and MOMA-2 positive cells in the mouse model. FXa stimulated Smad2/3 phosphorylation and MMP2 expression in aortic vascular smooth muscle cells (VSMC) in vitro. Expression of MMP2 in FXa-stimulated VSMC was downregulated in the presence of a PAR-2 but not a PAR-1 inhibitor. These findings suggest that FXa/FIIa inhibition limits aortic aneurysm and atherosclerosis severity due to down-regulation of vascular PAR-2-mediated Smad2/3 signalling and MMP2 expression. Inhibition of FXa/FIIa may be a potential therapy for limiting aortic aneurysm.
Collapse
|
70
|
Zhou D, Wan Z, Fan Y, Zhou J, Yuan Z. A combination of the neutrophil-to-lymphocyte ratio and the GRACE risk score better predicts PCI outcomes in Chinese Han patients with acute coronary syndrome. Anatol J Cardiol 2016; 15:995-1001. [PMID: 26663224 PMCID: PMC5368472 DOI: 10.5152/anatoljcardiol.2015.6174] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Objective: The aims of this study are to evaluate the relationship between the Global Registry of Acute Coronary Events (GRACE) risk score and neutrophil to lymphocyte ratio (NLR) and to determine whether a combination of these factors improves the predictive value for long-term cardiovascular events in Chinese Han patients with acute coronary syndrome (ACS). Methods: In this prospective, observational, and single-center study, NLRs (neutrophil count/lymphocyte count) were calculated from the complete blood count of 1050 patients with ACS, whereas GRACE risk scores were calculated from patients’ clinical parameters obtained on arrival at our hospital. Cox proportional hazards models were used to determine independent factors associated with cardiovascular events. Results: NLR was positively correlated with the GRACE risk score (r=0.66, p<0.001), and both the GRACE risk score (HR: 1.01; 95% CI: 1.01–1.02; p<0.001) and NLR (HR: 1.09; 95% CI: 1.06–1.14; p<0.001) independently predicted cardiovascular events. The area under the receiver operating characteristic (ROC) curve was 0.69 (95% CI: 0.64–0.72; p<0.001) when the GRACE score was calculated alone; however, it significantly increased (p<0.001) to 0.77 (95% CI: 0.74–0.80; p<0.001) when the GRACE score was combined with NLR. Conclusion: This study shows for the first time that NLR is positively associated with the GRACE risk score and demonstrates that a combination of these two factors may improve the predictive value for cardiovascular events in Chinese Han patients with ACS.
Collapse
Affiliation(s)
- Dong Zhou
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an-Xi'an-People's Republic of China.
| | | | | | | | | |
Collapse
|
71
|
Joshi MB, Baipadithaya G, Balakrishnan A, Hegde M, Vohra M, Ahamed R, Nagri SK, Ramachandra L, Satyamoorthy K. Elevated homocysteine levels in type 2 diabetes induce constitutive neutrophil extracellular traps. Sci Rep 2016; 6:36362. [PMID: 27811985 PMCID: PMC5095649 DOI: 10.1038/srep36362] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 10/13/2016] [Indexed: 12/26/2022] Open
Abstract
Constitutively active neutrophil extracellular traps (NETs) and elevated plasma homocysteine are independent risk factors for Type 2 Diabetes (T2D) associated vascular diseases. Here, we show robust NETosis due to elevated plasma homocysteine levels in T2D subjects and increased components of NETs such as neutrophil elastase and cell free DNA. Cooperative NETs formation was observed in neutrophils exposed to homocysteine, IL-6 and high glucose suggesting acute temporal changes tightly regulate constitutive NETosis. Homocysteine induced NETs by NADPH oxidase dependent and independent mechanisms. Constitutively higher levels of calcium and mitochondrial superoxides under hyperglycemic conditions were further elevated in response to homocysteine leading to accelerated NETosis. Homocysteine showed robust interaction between neutrophils and platelets by inducing platelet aggregation and NETosis in an interdependent manner. Our data demonstrates that homocysteine can alter innate immune function by promoting NETs formation and disturbs homeostasis between platelets and neutrophils which may lead to T2D associated vascular diseases.
Collapse
Affiliation(s)
| | | | | | - Mangala Hegde
- School of Life Sciences, Manipal University, Manipal, India
| | - Manik Vohra
- School of Life Sciences, Manipal University, Manipal, India
| | - Rayees Ahamed
- School of Life Sciences, Manipal University, Manipal, India
| | - Shivashankara K Nagri
- Department of Medicine, Kasturba Medical College, Manipal University, Manipal, India
| | | | | |
Collapse
|
72
|
Lijfering WM, Biedermann JS, Kruip MJ, Leebeek FW, Rosendaal FR, Cannegieter SC. Can we prevent venous thrombosis with statins: an epidemiologic review into mechanism and clinical utility. Expert Rev Hematol 2016; 9:1023-1030. [DOI: 10.1080/17474086.2016.1245137] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
73
|
Posma JJN, Posthuma JJ, Spronk HMH. Coagulation and non-coagulation effects of thrombin. J Thromb Haemost 2016; 14:1908-1916. [PMID: 27513692 DOI: 10.1111/jth.13441] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 07/08/2016] [Indexed: 01/06/2023]
Abstract
Thrombin is a multifunctional serine protease produced from prothrombin, and is a key regulator in hemostatic and non-hemostatic processes. It is the main effector protease in primary hemostasis by activating platelets, and plays a key role in secondary hemostasis. Besides its well-known functions in hemostasis, thrombin also plays a role in various non-hemostatic biological and pathophysiologic processes, predominantly mediated through activation of protease-activated receptors (PARs). Depending on several factors, such as the concentration of thrombin, the duration of activation, the location of PARs, the presence of coreceptors, and the formation of PAR heterodimers, activation of the receptor by thrombin can induce different cellular responses. Moreover, thrombin can have opposing effects in the same cell; it can induce both inflammatory and anti-inflammatory signals. Owing to the complexity of thrombin's signal transduction pathways, the exact mechanism behind the dichotomy of thrombin is yet still unknown. In this review, we highlight the hemostatic and non-hemostatic functions of thrombin, and specifically focus on the non-hemostatic dual role of thrombin under various conditions and in relation to cardiovascular disease.
Collapse
Affiliation(s)
- J J N Posma
- Departments of Internal Medicine and Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| | - J J Posthuma
- Departments of Internal Medicine and Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands
| | - H M H Spronk
- Departments of Internal Medicine and Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, the Netherlands.
| |
Collapse
|
74
|
Gulpen AJW, Ten Cate-Hoek AJ, Ten Cate H. Upstream versus downstream thrombin inhibition. Expert Rev Cardiovasc Ther 2016; 14:1273-1282. [DOI: 10.1080/14779072.2016.1224179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
75
|
Kim JB, Joung HJ, Lee JM, Woo JS, Kim WS, Kim KS, Lee KH, Kim W. Evaluation of the vascular protective effects of new oral anticoagulants in high-risk patients with atrial fibrillation (PREFER-AF): study protocol for a randomized controlled trial. Trials 2016; 17:422. [PMID: 27558002 PMCID: PMC4997652 DOI: 10.1186/s13063-016-1541-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 08/05/2016] [Indexed: 12/22/2022] Open
Abstract
Background Atrial fibrillation (AF) is known to be associated with several pathophysiological mechanisms including endothelial dysfunction of the heart and arterial vessels. Recent evidence suggests that new oral anticoagulant (NOAC) treatment may improve endothelial function and the inflammatory process involved in atherosclerosis in AF patients. This study is designed to determine the efficacy of NOAC therapy in the prevention of endothelial dysfunction and the progression of atherosclerosis of AF subjects. Method/design AF patients with a CHA2DS2-VASc score >2 and no previous history of overt coronary disease, severe peripheral arterial disease (PAD) or major stroke will be registered and randomly assigned either to the NOAC group (dabigatran or rivaroxaban) or the warfarin group in this prospective, randomized, 2-year follow-up study. Reactive hyperemia peripheral arterial tonometry (RH-PAT) measurements reflecting endothelial function will be conducted using the Endo-PAT2000 device. Left and right carotid intima-media thickness (IMT) will be measured at baseline, 12 months, and 24 months. The primary endpoint is defined as change in Reactive Hyperemia Index (RHI) at 12 months. Secondary endpoints included changes in the right and left maximum IMT of the common carotid artery (CCA) and internal carotid artery (ICA), the mean IMT of the CCA and ICA at 24 months, and 24-month cardiovascular events including cardiac death, stroke, acute myocardial infarction (AMI), overall cause of death, withdrawal of drug, or bleeding events. Discussion This is the first study to evaluate the efficacy of NOAC therapy for the prevention of endothelial dysfunction and progression of atherosclerosis in AF subjects. These findings are expected to expand the knowledge of NOAC pleotropic action in AF patients. Trial registration ClinicalTrials.gov: NCT02544932, registered on 7 September 2015.
Collapse
Affiliation(s)
- Jin-Bae Kim
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Hoegi-dong 1, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Hyun Jun Joung
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Hoegi-dong 1, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Jung Myung Lee
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Hoegi-dong 1, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Jong Shin Woo
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Hoegi-dong 1, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Woo-Shik Kim
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Hoegi-dong 1, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Kwon Sam Kim
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Hoegi-dong 1, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Kyung Hye Lee
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Hoegi-dong 1, Dongdaemun-gu, Seoul, 130-701, Republic of Korea
| | - Weon Kim
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Hoegi-dong 1, Dongdaemun-gu, Seoul, 130-701, Republic of Korea.
| |
Collapse
|
76
|
Affiliation(s)
- Hugo Ten Cate
- Departments of Internal Medicine and Biochemistry, Cardiovascular Research Institute Maastricht (CARIM) and Thrombosis Expertise Center, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - H Coenraad Hemker
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM) and Synapse, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
77
|
Affiliation(s)
- Nigel Mackman
- From the McAllister Heart Institute, Program in Thrombosis and Hemostasis, Division of Hematology/Oncology, Department of Medicine, University of North Carolina at Chapel Hill.
| |
Collapse
|
78
|
Grandoch M, Kohlmorgen C, Melchior-Becker A, Feldmann K, Homann S, Müller J, Kiene LS, Zeng-Brouwers J, Schmitz F, Nagy N, Polzin A, Gowert NS, Elvers M, Skroblin P, Yin X, Mayr M, Schaefer L, Tannock LR, Fischer JW. Loss of
Biglycan
Enhances Thrombin Generation in
Apolipoprotein E
-Deficient Mice. Arterioscler Thromb Vasc Biol 2016; 36:e41-50. [DOI: 10.1161/atvbaha.115.306973] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/17/2016] [Indexed: 11/16/2022]
Abstract
Objective—
Thrombin signaling promotes atherosclerosis by initiating inflammatory events indirectly through platelet activation and directly via protease-activated receptors. Therefore, endogenous thrombin inhibitors may be relevant modulators of atheroprogression and cardiovascular risk. In addition, endogenous thrombin inhibitors may affect the response to non–vitamin K-dependent oral anticoagulants. Here, the question was addressed whether the small leucine-rich proteoglycan biglycan acts as an endogenous thrombin inhibitor in atherosclerosis through activation of heparin cofactor II.
Approach and Results—
Biglycan concentrations were elevated in the plasma of patients with acute coronary syndrome and in male
Apolipoprotein E
-deficient (
ApoE
−/−
) mice. Biglycan was detected in the glycocalyx of capillaries and the subendothelial matrix of arterioles of
ApoE
−/−
mice and in atherosclerotic plaques. Thereby a vascular compartment is provided that may mediate the endothelial and subendothelial activation of heparin cofactor II through biglycan.
ApoE
and
Bgn
double-deficient (
ApoE
−/−
/Bgn
−/0
) mice showed higher activity of circulating thrombin, increased platelet activation and platelet adhesion in vivo, supporting a role of biglycan in balancing thrombin activity. Furthermore, concentrations of circulating cytokines and aortic macrophage content were elevated in
ApoE
−/−
/Bgn
−/0
mice, suggesting a proinflammatory phenotype. Elevated platelet activation and macrophage accumulation were reversed by treating
ApoE
−/−
/Bgn
−/0
mice with the thrombin inhibitor argatroban. Ultimately,
ApoE
−/−
/Bgn
−/0
mice developed aggravated atherosclerosis.
Conclusions—
The present results indicate that biglycan plays a previously unappreciated protective role during the progression of atherosclerosis by inhibiting thrombin activity, platelet activation, and finally macrophage-mediated plaque inflammation.
Collapse
Affiliation(s)
- Maria Grandoch
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Christina Kohlmorgen
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Ariane Melchior-Becker
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Kathrin Feldmann
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Susanne Homann
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Julia Müller
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Lena-Sophia Kiene
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Jinyang Zeng-Brouwers
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Friederike Schmitz
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Nadine Nagy
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Amin Polzin
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Nina S. Gowert
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Margitta Elvers
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Philipp Skroblin
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Xiaoke Yin
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Manuel Mayr
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Liliana Schaefer
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Lisa R. Tannock
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| | - Jens W. Fischer
- From the Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., J.W.F.); Cardiovascular Research Institute Düsseldorf (CARID), Universitätsklinikum der Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany (M.G., C.K., A.M.-B., K.F., S.H., J.M., L.-S.K., F.S., N.N., A.P., J.W.F.); Klinik für Kardiologie, Pneumologie und Angiologie,
| |
Collapse
|
79
|
Ellinghaus P, Perzborn E, Hauenschild P, Gerdes C, Heitmeier S, Visser M, Summer H, Laux V. Expression of pro-inflammatory genes in human endothelial cells: Comparison of rivaroxaban and dabigatran. Thromb Res 2016; 142:44-51. [PMID: 27131284 DOI: 10.1016/j.thromres.2016.04.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/31/2016] [Accepted: 04/12/2016] [Indexed: 01/29/2023]
Abstract
INTRODUCTION In addition to its central role in coagulation, thrombin is involved in non-hemostatic activities such as inflammation. Direct inhibition of thrombin activity (e.g. with dabigatran) or reducing its generation by inhibition of Factor Xa (e.g. with rivaroxaban) may therefore have anti-inflammatory effects. MATERIALS AND METHODS Microarray experiments were performed to identify transcriptome-wide changes in mRNA expression levels induced by thrombin in the presence and absence of the PAR-1 antagonist vorapaxar in primary human umbilical vein endothelial cells (HUVECs). On this basis, HUVECs were incubated with recalcified plasma, with or without rivaroxaban (0.3-3000nM), dabigatran (0.3-10,000nM), or vorapaxar (0.3-10nM). Expression levels of preselected pro-inflammatory genes were quantified by real-time PCR. RESULTS Vorapaxar abolished 67 of the 69 transcripts altered by more than twofold on addition of thrombin to HUVECs. ELAM-1, VCAM-1, ICAM-1, MCP-1, IL-8, CXCL1, and CXCL2 were among the genes most strongly induced by thrombin. Inflammatory gene expression after stimulation of thrombin generation was concentration-dependently suppressed by vorapaxar, dabigatran, and rivaroxaban. However, dabigatran at low concentrations (3-300nM) increased significantly the expression levels of CXCL1, CXCL2, IL-8, ELAM-1, MCP-1, and tissue factor. CONCLUSION In HUVECs, plasma-induced transcriptional changes are mediated by thrombin-induced PAR-1 activation. Rivaroxaban downregulated the expression of pro-inflammatory markers and tissue factor to a similar extent to dabigatran.
Collapse
Affiliation(s)
- Peter Ellinghaus
- Global Biomarkers Research, Bayer Pharma AG, Wuppertal, Germany.
| | - Elisabeth Perzborn
- Formerly Acute Care Diseases Research, Bayer Pharma AG, Wuppertal, Germany.
| | | | - Christoph Gerdes
- Acute Care Diseases Research, Bayer Pharma AG, Wuppertal, Germany.
| | - Stefan Heitmeier
- Acute Care Diseases Research, Bayer Pharma AG, Wuppertal, Germany.
| | - Mayken Visser
- Acute Care Diseases Research, Bayer Pharma AG, Wuppertal, Germany.
| | - Holger Summer
- Lead Generation & Optimization, Bayer Pharma AG, Wuppertal, Germany.
| | - Volker Laux
- Acute Care Diseases Research, Bayer Pharma AG, Wuppertal, Germany.
| |
Collapse
|
80
|
Spronk HMH, De Jong AM, Verheule S, De Boer HC, Maass AH, Lau DH, Rienstra M, van Hunnik A, Kuiper M, Lumeij S, Zeemering S, Linz D, Kamphuisen PW, Ten Cate H, Crijns HJ, Van Gelder IC, van Zonneveld AJ, Schotten U. Hypercoagulability causes atrial fibrosis and promotes atrial fibrillation. Eur Heart J 2016; 38:38-50. [PMID: 27071821 DOI: 10.1093/eurheartj/ehw119] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 12/21/2015] [Accepted: 03/02/2016] [Indexed: 01/09/2023] Open
Abstract
AIMS Atrial fibrillation (AF) produces a hypercoagulable state. Stimulation of protease-activated receptors by coagulation factors provokes pro-fibrotic, pro-hypertrophic, and pro-inflammatory responses in a variety of tissues. We studied the effects of thrombin on atrial fibroblasts and tested the hypothesis that hypercoagulability contributes to the development of a substrate for AF. METHODS AND RESULTS In isolated rat atrial fibroblasts, thrombin enhanced the phosphorylation of the pro-fibrotic signalling molecules Akt and Erk and increased the expression of transforming growth factor β1 (2.7-fold) and the pro-inflammatory factor monocyte chemoattractant protein-1 (6.1-fold). Thrombin also increased the incorporation of 3H-proline, suggesting enhanced collagen synthesis by fibroblasts (2.5-fold). All effects could be attenuated by the thrombin inhibitor dabigatran. In transgenic mice with a pro-coagulant phenotype (TMpro/pro), the inducibility of AF episodes lasting >1 s was higher (7 out of 12 vs. 1 out of 10 in wild type) and duration of AF episodes was longer compared with wild type mice (maximum episode duration 42.8 ± 68.4 vs. 0.23 ± 0.39 s). In six goats with persistent AF treated with nadroparin, targeting Factor Xa-mediated thrombin generation, the complexity of the AF substrate was less pronounced than in control animals (LA maximal activation time differences 23.3 ± 3.1 ms in control vs. 15.7 ± 2.1 ms in nadroparin, P < 0.05). In the treated animals, AF-induced α-smooth muscle actin expression was lower and endomysial fibrosis was less pronounced. CONCLUSION The hypercoagulable state during AF causes pro-fibrotic and pro-inflammatory responses in adult atrial fibroblasts. Hypercoagulability promotes the development of a substrate for AF in transgenic mice and in goats with persistent AF. In AF goats, nadroparin attenuates atrial fibrosis and the complexity of the AF substrate. Inhibition of coagulation may not only prevent strokes but also inhibit the development of a substrate for AF.
Collapse
Affiliation(s)
- Henri M H Spronk
- Department of Biochemistry, Maastricht University, Maastricht, The Netherlands.,Department of Internal Medicine, Maastricht University, Maastricht, The Netherlands
| | - Anne Margreet De Jong
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sander Verheule
- Department of Physiology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Hetty C De Boer
- Department of Nephrology and the Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, The Netherlands
| | - Alexander H Maass
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dennis H Lau
- Department of Physiology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Michiel Rienstra
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Arne van Hunnik
- Department of Physiology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Marion Kuiper
- Department of Physiology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Stijn Lumeij
- Department of Physiology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Stef Zeemering
- Department of Physiology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Dominik Linz
- Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, Homburg, Germany
| | - Pieter Willem Kamphuisen
- Department of Vascular Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hugo Ten Cate
- Department of Biochemistry, Maastricht University, Maastricht, The Netherlands.,Department of Internal Medicine, Maastricht University, Maastricht, The Netherlands
| | - Harry J Crijns
- Department of Cardiology, Academic Hospital Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Isabelle C Van Gelder
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anton Jan van Zonneveld
- Department of Nephrology and the Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, The Netherlands
| | - Ulrich Schotten
- Department of Physiology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
81
|
Shnerb Ganor R, Harats D, Schiby G, Gailani D, Levkovitz H, Avivi C, Tamarin I, Shaish A, Salomon O. Factor XI Deficiency Protects Against Atherogenesis in Apolipoprotein E/Factor XI Double Knockout Mice. Arterioscler Thromb Vasc Biol 2016; 36:475-81. [PMID: 26800563 DOI: 10.1161/atvbaha.115.306954] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 12/22/2015] [Indexed: 01/30/2023]
Abstract
OBJECTIVE Atherosclerosis and atherothrombosis are still major causes of mortality in the Western world, even after the widespread use of cholesterol-lowering medications. Recently, an association between local thrombin generation and atherosclerotic burden has been reported. Here, we studied the role of factor XI (FXI) deficiency in the process of atherosclerosis in mice. APPROACH AND RESULTS Apolipoprotein E/FXI double knockout mice, created for the first time in our laboratory. There was no difference in cholesterol levels or lipoprotein profiles between apolipoprotein E knockout and double knockout mice. Nevertheless, in 24-week-old double knockout mice, the atherosclerotic lesion area in the aortic sinus was reduced by 32% (P=0.004) in comparison with apolipoprotein E knockout mice. In 42-week-old double knockout mice, FXI deficiency inhibited atherosclerosis progression significantly in the aortic sinus (25% reduction, P=0.024) and in the aortic arch (49% reduction, P=0.028), with a prominent reduction of macrophage infiltration in the atherosclerotic lesions. CONCLUSIONS FXI deprivation was shown to slow down atherogenesis in mice. The results suggest that the development of atherosclerosis can be prevented by targeting FXI.
Collapse
Affiliation(s)
- Reut Shnerb Ganor
- From the The Bert W. Strassburger Lipid Center (R.S.G., D.H., H.L., A.S.) and Department of Pathology (G.S., C.A.), and Thrombosis Unit (R.S.G., I.T., O.S.), Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine (R.S.G., D.H., G.S., H.L., C.A., I.T., O.S.), Tel-Aviv University, Tel-Aviv, Israel; and Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN (D.G.)
| | - Dror Harats
- From the The Bert W. Strassburger Lipid Center (R.S.G., D.H., H.L., A.S.) and Department of Pathology (G.S., C.A.), and Thrombosis Unit (R.S.G., I.T., O.S.), Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine (R.S.G., D.H., G.S., H.L., C.A., I.T., O.S.), Tel-Aviv University, Tel-Aviv, Israel; and Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN (D.G.)
| | - Ginette Schiby
- From the The Bert W. Strassburger Lipid Center (R.S.G., D.H., H.L., A.S.) and Department of Pathology (G.S., C.A.), and Thrombosis Unit (R.S.G., I.T., O.S.), Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine (R.S.G., D.H., G.S., H.L., C.A., I.T., O.S.), Tel-Aviv University, Tel-Aviv, Israel; and Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN (D.G.)
| | - David Gailani
- From the The Bert W. Strassburger Lipid Center (R.S.G., D.H., H.L., A.S.) and Department of Pathology (G.S., C.A.), and Thrombosis Unit (R.S.G., I.T., O.S.), Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine (R.S.G., D.H., G.S., H.L., C.A., I.T., O.S.), Tel-Aviv University, Tel-Aviv, Israel; and Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN (D.G.)
| | - Hanna Levkovitz
- From the The Bert W. Strassburger Lipid Center (R.S.G., D.H., H.L., A.S.) and Department of Pathology (G.S., C.A.), and Thrombosis Unit (R.S.G., I.T., O.S.), Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine (R.S.G., D.H., G.S., H.L., C.A., I.T., O.S.), Tel-Aviv University, Tel-Aviv, Israel; and Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN (D.G.)
| | - Camila Avivi
- From the The Bert W. Strassburger Lipid Center (R.S.G., D.H., H.L., A.S.) and Department of Pathology (G.S., C.A.), and Thrombosis Unit (R.S.G., I.T., O.S.), Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine (R.S.G., D.H., G.S., H.L., C.A., I.T., O.S.), Tel-Aviv University, Tel-Aviv, Israel; and Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN (D.G.)
| | - Ilia Tamarin
- From the The Bert W. Strassburger Lipid Center (R.S.G., D.H., H.L., A.S.) and Department of Pathology (G.S., C.A.), and Thrombosis Unit (R.S.G., I.T., O.S.), Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine (R.S.G., D.H., G.S., H.L., C.A., I.T., O.S.), Tel-Aviv University, Tel-Aviv, Israel; and Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN (D.G.)
| | - Aviv Shaish
- From the The Bert W. Strassburger Lipid Center (R.S.G., D.H., H.L., A.S.) and Department of Pathology (G.S., C.A.), and Thrombosis Unit (R.S.G., I.T., O.S.), Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine (R.S.G., D.H., G.S., H.L., C.A., I.T., O.S.), Tel-Aviv University, Tel-Aviv, Israel; and Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN (D.G.)
| | - Ophira Salomon
- From the The Bert W. Strassburger Lipid Center (R.S.G., D.H., H.L., A.S.) and Department of Pathology (G.S., C.A.), and Thrombosis Unit (R.S.G., I.T., O.S.), Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine (R.S.G., D.H., G.S., H.L., C.A., I.T., O.S.), Tel-Aviv University, Tel-Aviv, Israel; and Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN (D.G.).
| |
Collapse
|
82
|
Shehab A, Elnour AA, Bhagavathula AS, Erkekoglu P, Hamad F, Al Nuaimi S, Al Shamsi A, Mukhtar I, Ali AbdElrazek AM, Al Suwaidi A, Mandil MA, Baraka M, Sadik A, Saraan K, Al Kalbani NM, Mahmood AA, Barqawi Y, Al Hajjar M, Shehab OA, Al Amoodi A, Asim S, Abdulla R, Giraud CS, Ahmed EM, Shaaban ZA, Eltayeb AEYA. Novel oral anticoagulants and the 73rd anniversary of historical warfarin. J Saudi Heart Assoc 2016; 28:31-45. [PMID: 26778903 PMCID: PMC4685209 DOI: 10.1016/j.jsha.2015.05.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 04/13/2015] [Accepted: 05/05/2015] [Indexed: 01/22/2023] Open
Affiliation(s)
- Abdulla Shehab
- Internal Medicine Department, College of Medicine and Health Sciences (CMHS), UAE University, United Arab Emirates
| | - Asim Ahmed Elnour
- Pharmacology Department, College of Medicine and Health Sciences, UAE University, United Arab Emirates
- Al Ain Hospital, Abu Dhabi Health Services, SEHA, United Arab Emirates
| | | | - Pınar Erkekoglu
- Department of Toxicology, Faculty of Pharmacy, Hacettepe University, Sihhiye 06100, Ankara, Turkey
| | - Farah Hamad
- Ajman University of Sciences and Technology, AJman, United Arab Emirates
| | - Saif Al Nuaimi
- Internal Medicine-Tawam Hospital, Abu Dhabi Health Services, SEHA, United Arab Emirates
| | - Ali Al Shamsi
- Internal Medicine-Tawam Hospital, Abu Dhabi Health Services, SEHA, United Arab Emirates
| | - Iman Mukhtar
- Al Ain Hospital, Abu Dhabi Health Services, SEHA, United Arab Emirates
| | | | - Aeshal Al Suwaidi
- Al Ain Hospital, Abu Dhabi Health Services, SEHA, United Arab Emirates
| | | | - Mohamed Baraka
- Department of Pharmacy Practice, College of Clinical Pharmacy-University of Dammam Eastern Province, Damman, Saudi Arabia
| | - Adel Sadik
- Al Ain Hospital, Abu Dhabi Health Services, SEHA, United Arab Emirates
| | - Khalid Saraan
- Internal Medicine, Abu Dhabi Rehabilitation Center, Abu Dhabi, United Arab Emirates
| | - Naama M.S. Al Kalbani
- Internal Medicine-Tawam Hospital, Abu Dhabi Health Services, SEHA, United Arab Emirates
| | - Alaa AbdulAziz Mahmood
- Pharmacology Department, College of Medicine and Health Sciences, UAE University, United Arab Emirates
| | - Yazan Barqawi
- Al Ain Hospital, Abu Dhabi Health Services, SEHA, United Arab Emirates
| | | | - Omer Abdulla Shehab
- Internal Medicine Department, College of Medicine and Health Sciences (CMHS), UAE University, United Arab Emirates
| | - Abdulla Al Amoodi
- Internal Medicine Department, College of Medicine and Health Sciences (CMHS), UAE University, United Arab Emirates
| | - Sahar Asim
- Ajman University of Sciences and Technology, AJman, United Arab Emirates
| | - Rauda Abdulla
- Internal Medicine Department, College of Medicine and Health Sciences (CMHS), UAE University, United Arab Emirates
| | - Cristina Sanches Giraud
- Universidade Federal de São João del-Rei, Campus Centro-Oeste Dona Lindu, Av. Sebastião Gonçalves Coelho, 400 Chanadour, Divinópolis, Brazil
| | - El Mutasim Ahmed
- Al Ain Hospital, Abu Dhabi Health Services, SEHA, United Arab Emirates
| | - Zohdi Abu Shaaban
- Al Ain Hospital, Abu Dhabi Health Services, SEHA, United Arab Emirates
| | | |
Collapse
|
83
|
Lau DH, Schotten U, Mahajan R, Antic NA, Hatem SN, Pathak RK, Hendriks JML, Kalman JM, Sanders P. Novel mechanisms in the pathogenesis of atrial fibrillation: practical applications. Eur Heart J 2015; 37:1573-81. [DOI: 10.1093/eurheartj/ehv375] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 07/21/2015] [Indexed: 12/21/2022] Open
|
84
|
|
85
|
Joseph P, Pare G, Wallentin L, Connolly S, Yusuf S, Wang J, Ezekowitz M, Eikelboom J, Siegbahn A, Reilly P, Themeles E, Oldgren J. Dabigatran etexilate and reduction in serum apolipoprotein B. Heart 2015; 102:57-62. [PMID: 26552755 DOI: 10.1136/heartjnl-2015-307586] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 08/19/2015] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Carboxylesterases, which convert dabigatran etexilate to its active form, dabigatran, have also been shown to influence lipoprotein metabolism, although any pleotropic effects of the drug based on this possible mechanism has not been evaluated. We examined the effects of dabigatran etexilate on serum lipoprotein markers in the Randomized Evaluation of Long-Term Anticoagulation Therapy (RE-LY) study. METHODS 2513 participants from the RE-LY randomised control trial with baseline and 3-month apolipoprotein B (ApoB) and apolipoprotein A1 (ApoA1) measurements were included. We prospectively compared the effects of dabigatran 110 mg twice daily, dabigatran 150 mg twice daily and warfarin on changes in ApoB and ApoA1 concentrations using a mixed model analysis. RESULTS From baseline to 3 months, a significant reduction in ApoB concentration was observed with low-dose dabigatran (-0.057 (95% CI -0.069 to -0.044) g/L, p<0.001) and high-dose dabigatran (-0.065 (95% CI -0.078 to -0.053) g/L, p<0.001) but not warfarin (-0.006 g/L (95% CI -0.018 to 0.007) g/L, p=0.40). Compared with warfarin, ApoB reduction was significantly greater with both doses of dabigatran (p<0.001 for both groups). Reductions in ApoA1 concentrations did not statistically differ with either dose of dabigatran when compared with warfarin. CONCLUSIONS Dabigatran is associated with a significant (∼7%) reduction in ApoB concentration, suggesting a novel effect of this drug on lipoprotein metabolism. Further studies are needed to determine the mechanism of this observed effect, and its impact on clinical outcomes.
Collapse
Affiliation(s)
- Philip Joseph
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Guillaume Pare
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Lars Wallentin
- Department of Medical Sciences, Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden
| | - Stuart Connolly
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Salim Yusuf
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Jia Wang
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | | | - John Eikelboom
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Agneta Siegbahn
- Department of Medical Sciences, Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden
| | - Paul Reilly
- Department of Clinical Development and Clinical Biostatistics, Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, USA
| | - Ellison Themeles
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Jonas Oldgren
- Department of Medical Sciences, Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
86
|
Chistiakov DA, Bobryshev YV, Orekhov AN. Neutrophil's weapons in atherosclerosis. Exp Mol Pathol 2015; 99:663-71. [PMID: 26551083 DOI: 10.1016/j.yexmp.2015.11.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 11/04/2015] [Indexed: 01/13/2023]
Abstract
Neutrophils are important components of immunity associated with inflammatory responses against a broad spectrum of pathogens. These cells could be rapidly activated by proinflammatory stimuli and migrate to the inflamed and infected sites where they release a variety of cytotoxic molecules with antimicrobial activity. Neutrophil antibacterial factors include extracellular proteases, redox enzymes, antimicrobial peptides, and small bioactive molecules. In resting neutrophils, these factors are stored in granules and released upon activation during degranulation. These factors could be also secreted in a neutrophil-derived microparticle-dependent fashion. Neutrophils exhibit a unique property to produce neutrophil extracellular traps (NETs) composed of decondensed chromatin and granular proteins to catch and kill bacteria. Neutrophil-released factors are efficient in inactivation and elimination of pathogens through oxidation-dependent or independent damage of bacterial cells, inactivation and neutralization of virulence factors and other mechanisms. However, in chronic atherosclerosis-associated inflammation, protective function of neutrophils could be impaired and misdirected against own cells. This could lead to deleterious effects and progressive vascular injury. In atherogenesis, a pathogenic role of neutrophils could be especially seen in early stages associated with endothelial dysfunction and induction of vascular inflammation and in late atherosclerosis associated with plaque rupture and atherothrombosis. Assuming a prominent impact of neutrophils in cardiovascular pathology, developing therapeutic strategies targeting neutrophil-specific antigens could have a promising clinical potential.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Molecular Genetic Diagnostics and Cell Biology, Division of Laboratory Medicine, Institute of Pediatrics, Research Center for Children's Health, 119991 Moscow, Russia
| | - Yuri V Bobryshev
- Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia; School of Medicine, University of Western Sydney, Campbelltown, NSW 2560, Australia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow 121609, Russia.
| | - Alexander N Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow 121609, Russia; Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow 125315, Russia; Department of Biophysics, Biological Faculty, Moscow State University, Moscow 119991, Russia
| |
Collapse
|
87
|
Preusch MR, Ieronimakis N, Wijelath ES, Cabbage S, Ricks J, Bea F, Reyes M, van Ryn J, Rosenfeld ME. Dabigatran etexilate retards the initiation and progression of atherosclerotic lesions and inhibits the expression of oncostatin M in apolipoprotein E-deficient mice. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:5203-11. [PMID: 26392754 PMCID: PMC4572747 DOI: 10.2147/dddt.s86969] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Objective Thrombin has multiple proatherogenic effects including platelet activation and the induction of inflammatory processes. Recently, the cytokine oncostatin M has been shown to have proinflammatory effects. This study was designed to investigate the effects of thrombin inhibition on the initiation and progression of atherosclerosis and on the expression of oncostatin M. Methods Apolipoprotein E-deficient mice at different ages were fed the thrombin inhibitor dabigatran etexilate. The mean lesion area was measured in the aortic sinus and in the innominate artery. CD45-positive cells within the aortic tissue were measured by flow cytometry. Oncostatin M expression was measured in the tissue sections by immunocytochemistry. Results Treatment with dabigatran etexilate resulted in a significant reduction of the mean area of atherosclerotic lesions in the aortic sinus in both the young mice (11,176±1,500 μm2 (control) versus 3,822±836 μm2 (dabigatran etexilate), P<0.05) and selectively in the older mice at 28 weeks (234,099±13,500 μm2 (control) versus 175,226±16,132 μm2 (dabigatran etexilate), P<0.05). There were also fewer CD45-positive cells within the aortas of the dabigatran-treated mice and enhanced NO production in endothelial cells pretreated with dabigatran. In addition, the expression of oncostatin M was reduced in the lesions of dabigatran etexilate-treated mice. Conclusion Inhibition of thrombin by dabigatran retards the development of early lesions and the progression of some established lesions in ApoE–/– mice. It improves endothelial function and retards macrophage accumulation within the vascular wall. Dabigatran also inhibits the expression of oncostatin M, and this suggests that oncostatin M may play a role in the initiation and progression of atherosclerosis.
Collapse
Affiliation(s)
- Michael R Preusch
- Department of Pathology, University of Washington, Seattle, WA, USA ; Department of Internal Medicine, University of Heidelberg, Heidelberg, Germany
| | | | - Errol S Wijelath
- Department of Surgery, University of Washington, Seattle, WA, USA
| | - Sara Cabbage
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Jerry Ricks
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Florian Bea
- Department of Internal Medicine, University of Heidelberg, Heidelberg, Germany
| | - Morayma Reyes
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Joanne van Ryn
- Department of CardioMetabolic Disease Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Michael E Rosenfeld
- Department of Pathology, University of Washington, Seattle, WA, USA ; Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| |
Collapse
|
88
|
Cartilage oligomeric matrix protein is a natural inhibitor of thrombin. Blood 2015; 126:905-14. [PMID: 26045608 DOI: 10.1182/blood-2015-01-621292] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 06/02/2015] [Indexed: 12/29/2022] Open
Abstract
Thrombin is an effector enzyme for hemostasis and thrombosis; however, endogenous regulators of thrombin remain elusive. Cartilage oligomeric matrix protein (COMP), a matricellular protein also known as thrombospondin-5, is essential for maintaining vascular homeostasis. Here, we asked whether COMP is involved in the process of blood coagulation. COMP deficiency shortened tail-bleeding and clotting time and accelerated ferric-chloride-induced thrombosis in mice. The absence of COMP had no effect on platelet count. In contrast, COMP specifically inhibited thrombin-induced platelet aggregation, activation, and retraction and the thrombin-mediated cleavage of fibrinogen. Furthermore, surface plasmon resonance analysis revealed direct thrombin-COMP binding (KD = 1.38 ± 0.24 μM). In particular, blockage of thrombin exosites with compounds specific for exosite I (hirudin and HD1 aptamer) or exosite II (heparin and HD22 aptamer) impaired the COMP-thrombin interaction, indicating a 2-site binding mechanism. Additionally, epidermal growth factor-like repeats (amino acids 84-261) were identified as a COMP binding site for thrombin. Moreover, COMP was expressed in and secreted by platelets. Using bone marrow transplantation and platelet transfusion to create chimeric mice, platelet-derived but not vessel-wall-derived COMP was demonstrated to inhibit coagulation. Taken together, COMP is an endogenous thrombin inhibitor and negative regulator of hemostasis and thrombosis.
Collapse
|
89
|
Mastenbroek TG, van Geffen JP, Heemskerk JWM, Cosemans JMEM. Acute and persistent platelet and coagulant activities in atherothrombosis. J Thromb Haemost 2015; 13 Suppl 1:S272-80. [PMID: 26149036 DOI: 10.1111/jth.12972] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The potential relevance of murine atherothrombosis models for understanding human disease has been debated in the past. Despite this, in the last decade, many thrombosis studies with atherogenic Apoe(-/-) mice have been performed, which provide novel insight into the molecular mechanisms by which platelet and coagulation processes accomplish acute thrombus formation after plaque disruption in vivo. Support for these mechanisms has come from whole blood flow perfusion studies over plaque material in vitro, which are also reviewed in this study. The main plaque-derived triggers for thrombus formation appear to be collagen and tissue factor, next to bioactive mediators such as prostaglandin E2. The atherothrombotic process relies on collagen- and ADP-receptor-induced platelet activation as well as on thrombin/fibrin generation via the extrinsic and intrinsic coagulation pathways. Less is known of the persistent effects of a thrombus on atherosclerosis progression, but evidence suggests roles herein of activated platelets and ongoing thrombin generation.
Collapse
Affiliation(s)
- T G Mastenbroek
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - J P van Geffen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - J W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - J M E M Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
90
|
Natorska J, Undas A. Blood coagulation and fibrinolysis in aortic valve stenosis: links with inflammation and calcification. Thromb Haemost 2015; 114:217-27. [PMID: 25809537 DOI: 10.1160/th14-10-0861] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 01/25/2015] [Indexed: 11/05/2022]
Abstract
Aortic valve stenosis (AS) increasingly afflicts our aging population. However, the pathobiology of the disease is still poorly understood and there is no effective pharmacotherapy for treating those at risk for clinical progression. The progression of AS involves complex inflammatory and fibroproliferative processes that resemble to some extent atherosclerosis. Accumulating evidence indicates that several coagulation proteins and its inhibitors, including tissue factor, tissue factor pathway inhibitor, prothrombin, factor XIII, von Willebrand factor, display increased expression within aortic stenotic valves, predominantly on macrophages and myofibroblasts around calcified areas. Systemic impaired fibrinolysis, along with increased plasma and valvular expression of plasminogen activator inhibitor-1, has also been observed in patients with AS in association with the severity of the disease. There is an extensive cross-talk between inflammation and coagulation in stenotic valve tissue which contributes to the calcification and mineralisation of the aortic valve leaflets. This review summarises the available data on blood coagulation and fibrinolysis in AS with the emphasis on their interactions with inflammation and calcification.
Collapse
Affiliation(s)
| | - A Undas
- Anetta Undas, Institute of Cardiology, Jagiellonian University School of Medicine, 80 Pradnicka St., 31-202 Cracow, Poland, Tel.: +48 12 6143004, Fax: +48 12 6143143, E-mail:
| |
Collapse
|
91
|
Sluimer JC, Gijbels MJ, Heeneman S. Detection of Intraplaque Hemorrhage in Mouse Atherosclerotic Lesions. Methods Mol Biol 2015; 1339:339-348. [PMID: 26445801 DOI: 10.1007/978-1-4939-2929-0_24] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Intraplaque hemorrhage is defined as the presence of fresh or lysed erythrocytes, iron deposits in macrophages, and/or a fibrin clot in an atherosclerotic plaque. These features can be detected by hematoxylin and eosin, Martius scarlet Blue, and Perl's iron histological stainings. It is noteworthy that intraplaque hemorrhage is only present in murine atherosclerotic plaques after additional interventions or additional genetic traits affecting matrix degradation or thrombosis. In this chapter, we describe methods to detect intraplaque hemorrhage in mouse atherosclerotic lesions.
Collapse
Affiliation(s)
- Judith C Sluimer
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, 5800, 6202 AZ, The Netherlands
| | - Marion J Gijbels
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, 5800, 6202 AZ, The Netherlands
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
- Department of Medical Biochemistry, Amsterdam Medical Center, Amsterdam, The Netherlands
| | - Sylvia Heeneman
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, 5800, 6202 AZ, The Netherlands.
| |
Collapse
|
92
|
Koch M, Zernecke A. The hemostatic system as a regulator of inflammation in atherosclerosis. IUBMB Life 2014; 66:735-44. [PMID: 25491152 DOI: 10.1002/iub.1333] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 11/19/2014] [Indexed: 11/07/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial vessel wall. As part of a tightly connected cross-talk between inflammation and coagulation, there is growing evidence that the coagulation system plays a pivotal role in the development and progression of atherosclerosis. We here discuss the presence of coagulation factors in atherosclerotic lesions and the overall effects of hypercoagulability and hypocoagulability on atherosclerotic lesion formation. Moreover, we focus on the unifying common pathway of coagulation, which can be initiated by the intrinsic and extrinsic pathway of coagulation, and discuss the functions of the coagulation factors FX, thrombin, and FXIII as regulators of inflammation in atherosclerosis. In particular, we review the non-hemostatic and immune-modulatory functions of these factors in endothelial and smooth muscle cells, as well as monocytes/macrophages, but also other cells, such as dendritic cells and T cells, that may control the inflammatory process of atherosclerosis. Their multiple roles in coagulation, but also their non-hemostatic functions in different cell types in inflammation and immunity, may harbor great potential for the development of novel therapeutic approaches for treating cardiovascular disease.
Collapse
Affiliation(s)
- Miriam Koch
- Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Würzburg, Germany
| | | |
Collapse
|
93
|
Schurgers LJ, Spronk HMH. Differential cellular effects of old and new oral anticoagulants: consequences to the genesis and progression of atherosclerosis. Thromb Haemost 2014; 112:909-17. [PMID: 25298033 DOI: 10.1160/th14-03-0268] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 09/16/2014] [Indexed: 01/06/2023]
Abstract
The main purpose of anticoagulants is to diminish fibrin formation, thereby decreasing the risk of venous or arterial thrombosis. Vitamin K antagonist have been used for many decades in order to achieve reduced thrombotic risk, despite major drawbacks of this class of drugs such as cumbersome dossing and monitoring of anticoagulant status. To overcome these drawbacks of VKA, new classes of anticoagulants have been developed including oral anticoagulants for direct inhibition of either thrombin or factor Xa, which can be administrated in a fixed dose without monitoring. Coagulation factors can activate cellular protease-activated receptors, thereby inducing cellular processes as inflammation, apoptosis, migration, and fibrosis. Therefore, inhibition of coagulation proteases not only attenuates fibrin formation, but may also influence pathophysiological processes like vascular calcification and atherosclerosis. Animal models revealed that VKA therapy induced both intima and media calcification and accelerated plaque vulnerability, whereas specific and direct inhibition of thrombin or factor Xa attenuated atherosclerosis. In this review we provide an overview of old and new oral anticoagulants, as well discuss potential pleiotropic effects with regard to calcification and atherosclerosis. Although translation from animal model to clinical patients seems difficult at first sight, effort should be made to fully understand the clinical implications of long-term oral anticoagulant therapy on vascular side effects.
Collapse
Affiliation(s)
- Leon J Schurgers
- Leon J. Schurgers, PhD, Department of Biochemistry, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands, Tel.: +31 433881681, Fax: +31 433884159, E-mail:
| | | |
Collapse
|
94
|
von Hundelshausen P, Schmitt MMN. Platelets and their chemokines in atherosclerosis-clinical applications. Front Physiol 2014; 5:294. [PMID: 25152735 PMCID: PMC4126210 DOI: 10.3389/fphys.2014.00294] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 07/22/2014] [Indexed: 12/22/2022] Open
Abstract
The concept of platelets as important players in the process of atherogenesis has become increasingly accepted due to accumulating experimental and clinical evidence. Despite the progress in understanding the molecular details of atherosclerosis, particularly by using animal models, the inflammatory and thrombotic roles of activated platelet s especially in the human system remain difficult to dissect, as often only the complications of atherosclerosis, i.e., stroke and myocardial infarction are definable but not the plaque burden. Platelet indices including platelet count and mean platelet volume (MPV) and soluble mediators released by activated platelets are associated with atherosclerosis. The chemokine CXCL4 has multiple atherogenic activities, e.g., altering the differentiation of T cells and macrophages by inhibiting neutrophil and monocyte apoptosis and by increasing the uptake of oxLDL and synergizing with CCL5. CCL5 is released and deposited on endothelium by activated platelets thereby triggering atherogenic monocyte recruitment, which can be attenuated by blocking the corresponding chemokine receptor CCR5. Atheroprotective and plaque stabilizing properties are attributed to CXCL12, which plays an important role in regenerative processes by attracting progenitor cells. Its release from luminal attached platelets accelerates endothelial healing after injury. Platelet surface molecules GPIIb/IIIa, GP1bα, P-selectin, JAM-A and the CD40/CD40L dyade are crucially involved in the interaction with endothelial cells, leukocytes and matrix molecules affecting atherogenesis. Beyond the effects on the arterial inflammatory infiltrate, platelets affect cholesterol metabolism by binding, modifying and endocytosing LDL particles via their scavenger receptors and contribute to the formation of lipid laden macrophages. Current medical therapies for the prevention of atherosclerotic therapies enable the elucidation of mechanisms linking platelets to inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Philipp von Hundelshausen
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University of Munich Munich, Germany ; German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance Munich, Germany
| | - Martin M N Schmitt
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University of Munich Munich, Germany
| |
Collapse
|
95
|
van Montfoort ML, Kuijpers MJE, Knaup VL, Bhanot S, Monia BP, Roelofs JJTH, Heemskerk JWM, Meijers JCM. Factor XI regulates pathological thrombus formation on acutely ruptured atherosclerotic plaques. Arterioscler Thromb Vasc Biol 2014; 34:1668-73. [PMID: 24947525 DOI: 10.1161/atvbaha.114.303209] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Coagulation factor XI is proposed as therapeutic target for anticoagulation. However, it is still unclear whether the antithrombotic properties of factor XI inhibitors influence atherosclerotic disease and atherothrombosis. Our aim is to investigate whether factor XI antisense oligonucleotides could prevent thrombus formation on acutely ruptured atherosclerotic plaques. APPROACH AND RESULTS Atherosclerotic plaques in the carotid arteries of Apoe(-/-) mice were acutely ruptured using ultrasound. The subsequent thrombus formation was visualized and quantified by intravital microscopy and immunohistochemistry. Mice were pretreated with either factor XI antisense or nonsense oligonucleotides (50 mg/kg) to lower factor XI plasma levels. A tail bleeding assay was used to determine the safety. On plaque rupture, initial platelet adhesion and platelet plug formation were not impaired in animals treated with factor XI antisense oligonucleotides. However, the ensuing thrombus formation and fibrin deposition were significantly lower after 5 to 10 minutes (P<0.05) in factor XI antisense oligonucleotide-treated animals without inducing a bleeding tendency. Furthermore, thrombi from antisense-treated animals were less stable than thrombi from placebo-treated animals. Moreover, macrophage infiltration and collagen deposition were lower in the carotid arteries of factor XI antisense-treated animals. No neutrophils were present. CONCLUSIONS Factor XI antisense oligonucleotides safely prevent thrombus formation on acutely ruptured atherosclerotic plaques in mice. Furthermore, perturbed carotid arteries from factor XI antisense-treated animals show a less severe inflammatory response.
Collapse
Affiliation(s)
- Maurits L van Montfoort
- From the Departments of Experimental Vascular Medicine (M.L.v.M., V.L.K., J.C.M.M.) and Pathology (J.J.T.H.R.), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (M.J.E.K., J.W.M.H.); Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc, Carlsbad, CA (S.B., B.P.M.); and Department of Plasma Proteins, Sanquin Research, Amsterdam, The Netherlands (J.C.M.M.)
| | - Marijke J E Kuijpers
- From the Departments of Experimental Vascular Medicine (M.L.v.M., V.L.K., J.C.M.M.) and Pathology (J.J.T.H.R.), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (M.J.E.K., J.W.M.H.); Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc, Carlsbad, CA (S.B., B.P.M.); and Department of Plasma Proteins, Sanquin Research, Amsterdam, The Netherlands (J.C.M.M.)
| | - Véronique L Knaup
- From the Departments of Experimental Vascular Medicine (M.L.v.M., V.L.K., J.C.M.M.) and Pathology (J.J.T.H.R.), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (M.J.E.K., J.W.M.H.); Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc, Carlsbad, CA (S.B., B.P.M.); and Department of Plasma Proteins, Sanquin Research, Amsterdam, The Netherlands (J.C.M.M.)
| | - Sanjay Bhanot
- From the Departments of Experimental Vascular Medicine (M.L.v.M., V.L.K., J.C.M.M.) and Pathology (J.J.T.H.R.), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (M.J.E.K., J.W.M.H.); Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc, Carlsbad, CA (S.B., B.P.M.); and Department of Plasma Proteins, Sanquin Research, Amsterdam, The Netherlands (J.C.M.M.)
| | - Brett P Monia
- From the Departments of Experimental Vascular Medicine (M.L.v.M., V.L.K., J.C.M.M.) and Pathology (J.J.T.H.R.), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (M.J.E.K., J.W.M.H.); Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc, Carlsbad, CA (S.B., B.P.M.); and Department of Plasma Proteins, Sanquin Research, Amsterdam, The Netherlands (J.C.M.M.)
| | - Joris J T H Roelofs
- From the Departments of Experimental Vascular Medicine (M.L.v.M., V.L.K., J.C.M.M.) and Pathology (J.J.T.H.R.), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (M.J.E.K., J.W.M.H.); Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc, Carlsbad, CA (S.B., B.P.M.); and Department of Plasma Proteins, Sanquin Research, Amsterdam, The Netherlands (J.C.M.M.)
| | - Johan W M Heemskerk
- From the Departments of Experimental Vascular Medicine (M.L.v.M., V.L.K., J.C.M.M.) and Pathology (J.J.T.H.R.), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (M.J.E.K., J.W.M.H.); Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc, Carlsbad, CA (S.B., B.P.M.); and Department of Plasma Proteins, Sanquin Research, Amsterdam, The Netherlands (J.C.M.M.)
| | - Joost C M Meijers
- From the Departments of Experimental Vascular Medicine (M.L.v.M., V.L.K., J.C.M.M.) and Pathology (J.J.T.H.R.), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands (M.J.E.K., J.W.M.H.); Department of Antisense Drug Discovery, Isis Pharmaceuticals Inc, Carlsbad, CA (S.B., B.P.M.); and Department of Plasma Proteins, Sanquin Research, Amsterdam, The Netherlands (J.C.M.M.).
| |
Collapse
|
96
|
Ten Cate H, Meade T. The Northwick Park Heart Study: evidence from the laboratory. J Thromb Haemost 2014; 12:587-92. [PMID: 24593861 DOI: 10.1111/jth.12545] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 02/25/2014] [Indexed: 01/12/2023]
Abstract
The Northwick Park Heart Study (NPHS) has shown associations of high plasma fibrinogen and factor VII (FVIIc) levels with the risk of death from coronary heart disease (CHD). The finding for fibrinogen has been confirmed in many other studies. Whereas one further study has found a similar prospective association for FVIIc, several have not. Experimental studies have demonstrated the impact that the coagulation activity of fibrinogen and FVIIc have on the progression and phenotype of atherosclerotic lesions. FVIIc-driven thrombin generation and fibrin formation within the vessel wall are important determinants of both plaque (in)stability and atherothrombosis. In blood, local concentrations of FVIIc and thrombin may be sufficient to allow interactions between these serine proteases and protease-activated receptors, to drive cellular inflammatory reactions that further promote these processes. Local fibrinogen concentrations dictate fibrin clot structure and resistance to fibrinolysis. Within the atherosclerotic plaque, coagulation reactions driven by proinflammatory stimuli may initially support lesion stability (as part of wound healing), but, with advanced inflammation, thrombin and fibrin generation diminish because of proteolytic activity contributing to plaque instability. The NPHS findings have proved controversial, but, in the light of current knowledge, a reappraisal of the importance of FVIIc and fibrinogen in atherosclerosis, atherothrombosis and CHD is justified. Hypercoagulability, reflected in turn by thrombin generation capacity, and local concentrations of coagulation proteins, including FVIIc and fibrinogen, is linked to plaque phenotype, and even minute local concentrations of fibrinogen and proteases such as FVIIc may affect thrombin generation capacity.
Collapse
Affiliation(s)
- H Ten Cate
- Laboratory of Clinical Thrombosis and Hemostasis, Department of Internal Medicine, Maastricht University Medical Center and Cardiovascular Research Institute, Maastricht, the Netherlands
| | | |
Collapse
|
97
|
Spronk HMH, Borissoff JI, ten Cate H. New insights into modulation of thrombin formation. Curr Atheroscler Rep 2014; 15:363. [PMID: 24026641 DOI: 10.1007/s11883-013-0363-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Thrombin is a pleiotropic enzyme that regulates hemostasis and nonhemostatic functions, including an array of actions within and on the vasculature. Physiologically, thrombin generation serves mainly to protect against thrombosis, but also to maintain vascular endothelial integrity. This protective effect is mediated in part through generation of anticoagulant enzymes, including activated protein C, formed on the action of thrombin on the endothelial receptor thrombomodulin. Partly, thrombin's vascular effects are effectuated through interaction with protease-activated receptors on various cell types. Pathophysiologically, downregulation and shedding of anticoagulant-acting receptors such as thrombomodulin and endothelial protein C receptor may contribute to a shift in activities of thrombin towards thrombogenic and proinflammatory actions. This shift may typically occur in the process of atherosclerosis, leading to a proatherogenic direction of the effects of thrombin. Therapeutically, the long-term inhibition of thrombin may create new ways of reducing atherosclerosis burden, altering the plaque phenotype.
Collapse
Affiliation(s)
- Henri M H Spronk
- Laboratory for Clinical Thrombosis and Haemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, P.O. Box 616, UNS50: Box8, 6200, MD, Maastricht, The Netherlands,
| | | | | |
Collapse
|
98
|
Gomes Quinderé AL, Benevides NMB, Carbone F, Mach F, Vuilleumier N, Montecucco F. Update on selective treatments targeting neutrophilic inflammation in atherogenesis and atherothrombosis. Thromb Haemost 2014; 111:634-646. [PMID: 24285257 DOI: 10.1160/th13-08-0712] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/28/2013] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is the most common pathological process underlying cardiovascular diseases. Current therapies are largely focused on alleviating hyperlipidaemia and preventing thrombotic complications, but do not completely eliminate risk of suffering recurrent acute ischaemic events. Specifically targeting the inflammatory processes may help to reduce this residual risk of major adverse cardiovascular events in atherosclerotic patients. The involvement of neutrophils in the pathophysiology of atherosclerosis is an emerging field, where evidence for their causal contribution during various stages of atherosclerosis is accumulating. Therefore, the identification of neutrophils as a potential therapeutic target may offer new therapeutic perspective to reduce the current atherosclerotic burden. This narrative review highlights the expanding role of neutrophils in atherogenesis and discusses on the potential treatment targeting neutrophil-related inflammation and associated atherosclerotic plaque vulnerability.
Collapse
Affiliation(s)
| | | | | | | | | | - Fabrizio Montecucco
- Fabrizio Montecucco, MD, PhD, Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, 4 rue Gabrielle-Perret-Gentil, 1205 Geneva, Switzerland, Tel: +41 22 38 27 238, Fax: +41 22 38 27 245, E mail:
| |
Collapse
|
99
|
Abstract
Understanding the pathophysiology of atherogenesis and the progression of atherosclerosis have been major goals of cardiovascular research during the previous decades. However, the complex molecular and cellular mechanisms underlying plaque destabilization remain largely obscure. Here, we review how lesional cells undergo cell death and how failed clearance exacerbates necrotic core formation. Advanced atherosclerotic lesions are further weakened by the pronounced local activity of matrix-degrading proteases as well as immature neovessels sprouting into the lesion. To stimulate translation of the current knowledge of molecular mechanisms of plaque destabilization into clinical studies, we further summarize available animal models of plaque destabilization. Based on the molecular mechanisms leading to plaque instability, we outline the current status of clinical and preclinical trials to induce plaque stability with a focus on induction of dead cell clearance, inhibition of protease activity, and dampening of inflammatory cell recruitment.
Collapse
|
100
|
Spronk HMH, de Jong AM, Crijns HJ, Schotten U, Van Gelder IC, ten Cate H. Pleiotropic effects of factor Xa and thrombin: what to expect from novel anticoagulants. Cardiovasc Res 2014; 101:344-51. [DOI: 10.1093/cvr/cvt343] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|