1
|
Wu Z, Sun J, Liao Z, Sun T, Huang L, Qiao J, Ling C, Chen C, Zhang B, Wang H. Activation of PAR1 contributes to ferroptosis of Schwann cells and inhibits regeneration of myelin sheath after sciatic nerve crush injury in rats via Hippo-YAP/ACSL4 pathway. Exp Neurol 2025; 384:115053. [PMID: 39542339 DOI: 10.1016/j.expneurol.2024.115053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/06/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE Peripheral nerve injury (PNI) is characterized by high incidence and sequela rate. Recently, there was increasing evidence that has shown ferroptosis may impede functional recovery. Our objective is to explore the novel mechanism that regulates ferroptosis after PNI. METHODS LC-MS/MS proteomics was used to explore the possible differential signals, while PCR array was performed to investigate the differential factors. Besides, we also tried to activate or inhibit the key factors and then observe the level of ferroptosis. Regeneration of myelin sheath was finally examined in vivo via transmission electron microscopy. RESULTS Proteomics analysis suggested coagulation signal was activated after sciatic nerve crush injury, in which high expression of F2 (encoding thrombin) and F2r (encoding PAR1) were observed. Both thrombin and PAR1-targeted activator TRAP6 can induce ferroptosis in RSC96 cells, which can be rescued by Vorapaxar (PAR1 targeted inhibitor) in vitro. Further PCR array revealed that activation of PAR1 induced ferroptosis in RSC96 cells by increasing expression of YAP and ACSL4. Immunofluorescence of sciatic nerve confirmed that the expression of YAP and ACSL4 were simultaneously reduced after PAR1 inhibition, which may contribute to myelin regeneration after injury in SD rats. CONCLUSION Inhibition of PAR1 can relieve ferroptosis after sciatic nerve crush injury in SD rats through Hippo-YAP/ACSL4 pathway, thereby regulating myelin regeneration after injury. In summary, PAR1/Hippo-YAP/ACSL4 pathway may be a promising therapeutic target for promoting functional recovery post-sciatic crush injury.
Collapse
Affiliation(s)
- Zhimin Wu
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Jun Sun
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Zhi Liao
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Tao Sun
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Lixin Huang
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Jia Qiao
- Department of Rehabilitation Medicine, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Cong Ling
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Chuan Chen
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Baoyu Zhang
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| | - Hui Wang
- Department of Neurosurgery, the Third Affiliated Hospital, Sun Yat-Sen University, 600 Tian He Road, Tian He District, Guangzhou, Guangdong 510630, China.
| |
Collapse
|
2
|
O'Brien OM, Tremble SM, Kropf A, Cipolla MJ. Thrombin in Pregnancy and Preeclampsia: Expression, Localization, and Vasoactivity in Brain and Microvessels From Rats. J Cardiovasc Pharmacol 2024; 84:250-260. [PMID: 38922586 PMCID: PMC11402023 DOI: 10.1097/fjc.0000000000001579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/12/2024] [Indexed: 06/27/2024]
Abstract
ABSTRACT Thrombin is a coagulation factor increased in pregnancy and further increased in preeclampsia (PE), a hypertensive disorder. Thrombin is also expressed in the brain and may have a nonhemostatic role. We characterized thrombin expression and vasoactivity in brain cerebral parenchymal arterioles (PAs) in rat models of pregnancy and PE. PAs were isolated and pressurized from nonpregnant (NP) and late-pregnant (LP) rats and rats with experimental preeclampsia (ePE). Reactivity to thrombin (1-50 U/mL) was measured in the absence and presence of inhibition of cyclooxygenase and nitric oxide synthase. Plasma levels of prothrombin, thrombin-antithrombin (TAT), tissue plasminogen activator, and plasminogen activator inhibitor-1 (PAI-1) and cerebrospinal fluid levels of TAT were compared using enzyme-linked immunosorbent assay. Expression of protease-activated receptor types 1 and 2 in PAs were measured by Western blot and immunohistochemistry. Neuronal thrombin expression was quantified in brains from all groups by immunohistochemistry. Prothrombin and TAT were elevated in ePE plasma compared with NP and LP. TAT was detected in cerebrospinal fluid from all groups and significantly elevated in LP (NP: 0.137 ± 0.014 ng/mL, LP: 0.241 ± 0.015 ng/mL, ePE: 0.192 ± 0.028 ng/mL; P < 0.05). Thrombin caused modest vasoconstriction in PAs from all groups regardless of cyclooxygenase or nitric oxide synthase inhibition. PAR1 and PAR2 were found in PAs from all groups colocalized to smooth muscle. Thrombin expression in central neurons was decreased in both LP and ePE groups compared with NP. These findings suggest a role for thrombin and other hemostatic changes during pregnancy and PE beyond coagulation.
Collapse
Affiliation(s)
- Olivia M O'Brien
- Department of Electrical and Biomedical Engineering, University of Vermont College of Engineering and Mathematical Sciences, Burlington, VT
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT
| | - Sarah M Tremble
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT
| | - Ari Kropf
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT
| | - Marilyn J Cipolla
- Department of Electrical and Biomedical Engineering, University of Vermont College of Engineering and Mathematical Sciences, Burlington, VT
- Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont Larner College of Medicine, Burlington, VT; and
- Department of Pharmacology, University of Vermont Larner College of Medicine, Burlington, VT
| |
Collapse
|
3
|
Iba T, Levi M, Levy JH. Intracellular communication and immunothrombosis in sepsis. J Thromb Haemost 2022; 20:2475-2484. [PMID: 35979601 PMCID: PMC9804233 DOI: 10.1111/jth.15852] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/22/2022] [Accepted: 08/10/2022] [Indexed: 01/05/2023]
Abstract
Inflammation and coagulation are the critical responses to infection that include leukocytes, platelets, and vascular endothelial cells responding in concert to eradicate the invading pathogen. In sepsis, a variety of cell surface receptors, including toll-like receptors, Fcγ-receptors, G-protein-coupled receptors, and adhesion receptors, detect the pathogens and elicit thromboinflammatory responses. Concurrently, the molecular patterns released from host damaged cells accelerate the immune responses through binding to the same pattern recognition receptors. Cytokines, chemokines, and extracellular vesicles are important mediators for amplifying the responses to distant cells as part of the systemic response to infections. At the same time, cells communicate with each other via direct contact, adhesion molecules, paracrine mediators, and tunneling nanotubes, which are important for regulating inflammation and thrombus formation. Despite increasing attention to immunothrombosis in sepsis, these close communication systems are less understood but play a critical role in host defense mechanisms. In this review, cellular activation and direct intercellular communication systems in sepsis with a focus on the coagulation response will be considered.
Collapse
Affiliation(s)
- Toshiaki Iba
- Department of Emergency and Disaster MedicineJuntendo University Graduate School of MedicineTokyoJapan
| | - Marcel Levi
- Department of Vascular Medicine, Amsterdam University Medical Center, the Netherlands and Department of MedicineUniversity College London Hospitals NHS Foundation Trust, and Cardio‐metabolic Programme‐NIHR UCLH/UCL BRC LondonLondonUK
| | - Jerrold H. Levy
- Department of Anesthesiology, Critical Care, and SurgeryDuke University School of MedicineDurhamNorth CarolinaUSA
| |
Collapse
|
4
|
ten Cate H, Guzik TJ, Eikelboom J, Spronk HMH. Pleiotropic actions of factor Xa inhibition in cardiovascular prevention: mechanistic insights and implications for anti-thrombotic treatment. Cardiovasc Res 2021; 117:2030-2044. [PMID: 32931586 PMCID: PMC8318102 DOI: 10.1093/cvr/cvaa263] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/10/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease in which atherothrombotic complications lead to cardiovascular morbidity and mortality. At advanced stages, myocardial infarction, ischaemic stroke, and peripheral artery disease, including major adverse limb events, are caused either by acute occlusive atherothrombosis or by thromboembolism. Endothelial dysfunction, vascular smooth muscle cell activation, and vascular inflammation are essential in the development of acute cardiovascular events. Effects of the coagulation system on vascular biology extend beyond thrombosis. Under physiological conditions, coagulation proteases in blood are pivotal in maintaining haemostasis and vascular integrity. Under pathological conditions, including atherosclerosis, the same coagulation proteases (including factor Xa, factor VIIa, and thrombin) become drivers of atherothrombosis, working in concert with platelets and vessel wall components. While initially atherothrombosis was attributed primarily to platelets, recent advances indicate the critical role of fibrin clot and plasma coagulation factors. Mechanisms of atherothrombosis and hypercoagulability vary depending on plaque erosion or plaque rupture. In addition to contributing to thrombus formation, factor Xa and thrombin can affect endothelial dysfunction, oxidative stress, vascular smooth muscle cell function as well as immune cell activation and vascular inflammation. By these mechanisms, they promote atherosclerosis and contribute to plaque instability. In this review, we first discuss the postulated vasoprotective mechanisms of protease-activated receptor signalling induced by coagulation enzymes under physiological conditions. Next, we discuss preclinical studies linking coagulation with endothelial cell dysfunction, thromboinflammation, and atherogenesis. Understanding these mechanisms is pivotal for the introduction of novel strategies in cardiovascular prevention and therapy. We therefore translate these findings to clinical studies of direct oral anticoagulant drugs and discuss the potential relevance of dual pathway inhibition for atherothrombosis prevention and vascular protection.
Collapse
Affiliation(s)
- Hugo ten Cate
- Department of Internal Medicine, Thrombosis Expertise Center, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, PO Box 616, 6200 MD, Maastricht, The Netherlands
- Department of Biochemistry, Thrombosis Expertise Center, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, PO Box 616, 6200 MD, Maastricht, The Netherlands
| | - Tomasz J Guzik
- Institute of Cardiovascular & Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, Glasgow, UK
- Department of Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland
| | - John Eikelboom
- Population Health Research Institute, Hamilton General Hospital and McMaster University, Hamilton, L8L 2x2, ON, Canada
| | - Henri M H Spronk
- Department of Internal Medicine, Thrombosis Expertise Center, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, PO Box 616, 6200 MD, Maastricht, The Netherlands
- Department of Biochemistry, Thrombosis Expertise Center, Cardiovascular Research Institute Maastricht, Maastricht University, Universiteitssingel 50, PO Box 616, 6200 MD, Maastricht, The Netherlands
| |
Collapse
|
5
|
Ekholm M, Kahan T. The Impact of the Renin-Angiotensin-Aldosterone System on Inflammation, Coagulation, and Atherothrombotic Complications, and to Aggravated COVID-19. Front Pharmacol 2021; 12:640185. [PMID: 34220496 PMCID: PMC8245685 DOI: 10.3389/fphar.2021.640185] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 06/07/2021] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis is considered a disease caused by a chronic inflammation, associated with endothelial dysfunction, and several mediators of inflammation are up-regulated in subjects with atherosclerotic disease. Healthy, intact endothelium exhibits an antithrombotic, protective surface between the vascular lumen and vascular smooth muscle cells in the vessel wall. Oxidative stress is an imbalance between anti- and prooxidants, with a subsequent increase of reactive oxygen species, leading to tissue damage. The renin-angiotensin-aldosterone system is of vital importance in the pathobiology of vascular disease. Convincing data indicate that angiotensin II accelerates hypertension and augments the production of reactive oxygen species. This leads to the generation of a proinflammatory phenotype in human endothelial and vascular smooth muscle cells by the up-regulation of adhesion molecules, chemokines and cytokines. In addition, angiotensin II also seems to increase thrombin generation, possibly via a direct impact on tissue factor. However, the mechanism of cross-talk between inflammation and haemostasis can also contribute to prothrombotic states in inflammatory environments. Thus, blocking of the renin-angiotensin-aldosterone system might be an approach to reduce both inflammatory and thrombotic complications in high-risk patients. During COVID-19, the renin-angiotensin-aldosterone system may be activated. The levels of angiotensin II could contribute to the ongoing inflammation, which might result in a cytokine storm, a complication that significantly impairs prognosis. At the outbreak of COVID-19 concerns were raised about the use of angiotensin converting enzyme inhibitors and angiotensin receptor blocker drugs in patients with COVID-19 and hypertension or other cardiovascular comorbidities. However, the present evidence is in favor of continuing to use of these drugs. Based on experimental evidence, blocking the renin-angiotensin-aldosterone system might even exert a potentially protective influence in the setting of COVID-19.
Collapse
Affiliation(s)
- M Ekholm
- Karolinska Institutet, Department of Clinical Sciences, Danderyd Hospital, Division of Cardiovascular Medicine, Stockholm, Sweden
| | - T Kahan
- Karolinska Institutet, Department of Clinical Sciences, Danderyd Hospital, Division of Cardiovascular Medicine, Stockholm, Sweden
| |
Collapse
|
6
|
Fujii N, McNeely BD, Zhang SY, Abdellaoui YC, Danquah MO, Kenny GP. Activation of protease-activated receptor 2 mediates cutaneous vasodilatation but not sweating: roles of nitric oxide synthase and cyclo-oxygenase. Exp Physiol 2018; 102:265-272. [PMID: 27981668 DOI: 10.1113/ep086092] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/05/2016] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? Protease-activated receptor 2 (PAR2) is located in the endothelial cells of skin vessels and eccrine sweat glands. However, a functional role of PAR2 in the control of cutaneous blood flow and sweating remains to be assessed in humans in vivo. What is the main finding and its importance? Our results demonstrate that in normothermic resting humans in vivo, activation of PAR2 elicits cutaneous vasodilatation partly through nitric oxide synthase-dependent mechanisms, but does not mediate sweating. These results provide important new insights into the physiological significance of PAR2 in human skin. Protease-activated receptor 2 (PAR2) is present in human skin, including keratinocytes, endothelial cells of skin microvessels and eccrine sweat glands. However, whether PAR2 contributes functionally to the regulation of cutaneous blood flow and sweating remains entirely unclear in humans in vivo. We hypothesized that activation of PAR2 directly stimulates cutaneous vasodilatation and sweating via actions of nitric oxide synthase (NOS) and cyclo-oxygenase (COX). In 12 physically active young men (29 ± 5 years old), cutaneous vascular conductance (CVC) and sweat rate were measured at four intradermal microdialysis forearm skin sites that were treated with the following: (i) lactated Ringer's solution (control); (ii) 10 mm NG -nitro-l-arginine (NOS inhibitor); (iii) 10 mm ketorolac (COX inhibitor); or (iv) a combination of both inhibitors. At all sites, a PAR2 agonist (SLIGKV-NH2 ) was co-administered in a dose-dependent fashion (0.06, 0.18, 0.55, 1.66 and 5 mm, each for 25 min). The highest dose of SLIGKV-NH2 (5 mm) increased CVC from baseline at the control site (P ≤ 0.05). This increase in CVC associated with PAR2 activation was attenuated by NOS inhibition regardless of the presence or absence of simultaneous COX inhibition (both P ≤ 0.05). However, COX inhibition alone did not affect the PAR2-mediated increase in CVC (P > 0.05). No increase in sweat rate was measured at any administered dose of SLIGKV-NH2 (all P > 0.05). We show that in normothermic resting humans in vivo, PAR2 activation does not increase sweat rate, whereas it does modulate cutaneous vasodilatation through NOS-dependent mechanisms.
Collapse
Affiliation(s)
- Naoto Fujii
- Human and Environmental Physiology Research Unit, University of Ottawa, Ottawa, Ontario, Canada
| | - Brendan D McNeely
- Human and Environmental Physiology Research Unit, University of Ottawa, Ottawa, Ontario, Canada
| | - Sarah Y Zhang
- Human and Environmental Physiology Research Unit, University of Ottawa, Ottawa, Ontario, Canada
| | - Yasmine C Abdellaoui
- Human and Environmental Physiology Research Unit, University of Ottawa, Ottawa, Ontario, Canada
| | - Mercy O Danquah
- Human and Environmental Physiology Research Unit, University of Ottawa, Ottawa, Ontario, Canada
| | - Glen P Kenny
- Human and Environmental Physiology Research Unit, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
7
|
Characterization and Functions of Protease-Activated Receptor 2 in Obesity, Diabetes, and Metabolic Syndrome: A Systematic Review. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3130496. [PMID: 27006943 PMCID: PMC4781943 DOI: 10.1155/2016/3130496] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/26/2016] [Indexed: 12/16/2022]
Abstract
Proteinase-activated receptor 2 (PAR2) is a cell surface receptor activated by serine proteinases or specific synthetic compounds. Interest in PAR2 as a pharmaceutical target for various diseases is increasing. Here we asked two questions relevant to endothelial dysfunction and diabetes: How is PAR2 function affected in blood vessels? What role does PAR2 have in promoting obesity, diabetes, and/or metabolic syndrome, specifically via the endothelium and adipose tissues? We conducted a systematic review of the published literature in PubMed and Scopus (July 2015; search terms: par2, par-2, f2lr1, adipose, obesity, diabetes, and metabolic syndrome). Seven studies focused on PAR2 and vascular function. The obesity, diabetes, or metabolic syndrome animal models differed amongst studies, but each reported that PAR2-mediated vasodilator actions were preserved in the face of endothelial dysfunction. The remaining studies focused on nonvascular functions and provided evidence supporting the concept that PAR2 activation promoted obesity. Key studies showed that PAR2 activation regulated cellular metabolism, and PAR2 antagonists inhibited adipose gain and metabolic dysfunction in rats. We conclude that PAR2 antagonists for treatment of obesity indeed show early promise as a therapeutic strategy; however, endothelial-specific PAR2 functions, which may offset mechanisms that produce vascular dysfunction in diabetes, warrant additional study.
Collapse
|
8
|
Upregulation of Dickkopf1 by oscillatory shear stress accelerates atherogenesis. J Mol Med (Berl) 2015; 94:431-41. [PMID: 26607269 DOI: 10.1007/s00109-015-1369-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 10/12/2015] [Accepted: 11/18/2015] [Indexed: 12/16/2022]
Abstract
UNLABELLED Numerous clinical studies have highlighted the pivotal role Dickkopf (DKK) 1 plays in atherosclerosis, but the underlying mechanisms remain unknown. The present study was designed to explore the contribution of DKK1 to the development of atherosclerosis under oscillatory shear stress. Oscillatory shear stress applied to endothelial cells induced DKK1 expression, which peaked at 6 h. siRNA knockdown or silencing DKK1 by lentiviral gene delivery counteracted the increased monocyte adhesion and impaired endothelial tight junction induced by oscillatory shear stress, thereby attenuating atherogenesis in ApoE-/- mice. As well, activation of endothelial proteinase-activated receptor 1 (PAR1) and its downstream transcription factor, cAMP response element-binding protein (CREB), was critical to the increased expression of DKK1 under oscillatory shear stress. We provide evidence that DKK1 contributes to the development of atherosclerosis under conditions of oscillatory shear stress. A better understanding of the role played by DKK1 in atherogenesis may provide clinicians with opportunities to prevent atherosclerosis. KEY MESSAGE Disturbed oscillatory flow increases DKK1 expression. DKK1 knockdown attenuates OSS-induced monocyte adhesion and endothelial impairment. Genetic silencing of DKK1 limits atherogenesis in ApoE-/- mice. Activation of the PAR1/CREB pathway contributes to the upregulation of DKK1 via OSS. DKK1 is a promising candidate with respect to the treatment of atherosclerosis.
Collapse
|
9
|
Inverse association of the endogenous thrombin potential (ETP) with cardiovascular death: The Ludwigshafen Risk and Cardiovascular Health (LURIC) study. Int J Cardiol 2014; 176:139-44. [DOI: 10.1016/j.ijcard.2014.07.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 07/05/2014] [Indexed: 11/18/2022]
|
10
|
Alberelli MA, De Candia E. Functional role of protease activated receptors in vascular biology. Vascul Pharmacol 2014; 62:72-81. [PMID: 24924409 DOI: 10.1016/j.vph.2014.06.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 05/30/2014] [Accepted: 06/01/2014] [Indexed: 01/22/2023]
Abstract
Protease activated receptors (PARs) are a small family of G protein-coupled receptors (GPCR) mediating the cellular effects of some proteases of the coagulation system, such as thrombin, or other proteases, such as trypsin or metalloproteinase 1. As the prototype of PARs, PAR1 is a seven transmembrane GPCR that, upon cleavage by thrombin, unmasks a new amino-terminus able to bind intramolecularly to PAR1 itself thus inducing signaling. In the vascular system, thrombin and other proteases of the coagulation-fibrinolysis system, such as plasmin, factor VIIa and factor Xa, activated protein C, are considered physiologically relevant agonists, and PARs appear to largely account for the cellular effects of these enzymes. In the vasculature, PARs are expressed on platelets, endothelial cells (ECs) and vascular smooth muscle cells (VSMCs). In the vessel wall, under physiological conditions, PARs are mainly expressed in ECs and participate in the regulation of vascular tone, by inducing endothelium-dependent relaxation. PAR activation on ECs promotes conversion of these cells into a proinflammatory phenotype, causes increase of vascular permeability, and the exposure/secretion of proteins and cytokines mediating the local accumulation of platelets and leukocytes. These effects contribute to the vascular consequences of sepsis and of diseases such as acute lung injury and acute respiratory distress syndrome. In normal arteries PARs are to a much lesser amount expressed on VSMCs. However, in conditions associated with endothelial dysfunction, PARs mediate contraction, proliferation, migration, hypertrophy of VSMCs and their production of extracellular matrix, thereby contributing to the pathophysiology of atherosclerosis and hypertension. Inhibition of protease-PAR interaction might thus become a potential therapeutic target in various vascular diseases.
Collapse
Affiliation(s)
- Maria Adele Alberelli
- Hemostasis and Thrombosis Unit, Department of Internal Medicine, Agostino Gemelli Hospital School of Medicine, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Erica De Candia
- Hemostasis and Thrombosis Unit, Department of Internal Medicine, Agostino Gemelli Hospital School of Medicine, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
11
|
Abstract
Currently available antiplatelet agents have shown improved short- and long-term clinical outcomes but are associated with increased bleeding risk, and the rates of recurrent ischemic events still remain high. Selective inhibition of protease-activated receptor-1 for thrombin represents a potential novel strategy to reduce ischemic events without increasing the risk of bleeding. Two protease-activated receptor-1 antagonists are currently being evaluated in clinical trials: SCH 530348 and E5555. Results of phase II trials have shown that SCH 530348, when added to standard antiplatelet therapy, was well tolerated and not associated with increased bleeding risk. Two large-scale phase III trials assessing the efficacy of SCH 530348 in addition to the standard of care are currently ongoing. This review provides an outline of the current status of understanding on platelet thrombin-receptor antagonist SCH 530348, focusing on its pharmacologic properties and clinical development.
Collapse
|
12
|
Hughes KH, Wijekoon EP, Valcour JE, Chia EW, McGuire JJ. Effects of chronic in-vivo treatments with protease-activated receptor 2 agonist on endothelium function and blood pressures in mice. Can J Physiol Pharmacol 2013; 91:295-305. [DOI: 10.1139/cjpp-2012-0266] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Short-term treatments with protease-activated receptor 2-activating peptides (PAR2-AP) induce endothelium-dependent vasodilation and decrease blood pressure. In this study, we tested the effect of chronic in-vivo treatment with PAR2-AP on the blood pressure and endothelium function of mice. Male PAR2 wild-type (WT) and par2-deficient (KO) mice received subcutaneous infusions of either saline, low (PAR2-LD), or high (PAR2-HD) doses of 2-furoyl-LIGRLO-amide for 1 or 2 weeks. In each treatment group, endothelium function was assessed in isolated arteries. Blood pressure, heart rate, and locomotor activity were recorded by radiotelemetry, and levels of tumour nercrosis factor α (TNF-α) and interkeukin 1β (IL-1β) were measured in plasma samples by ELISA. The relaxation of WT aortas and mesenteric arteries induced by PAR2-AP was decreased by PAR2-LD and PAR2-HD. In mesenteric arteries, PAR2-LD and PAR2-HD decreased the relaxation induced by acetylcholine, but not by nitroprusside; in aortas, PAR2-LD and PAR2-HD caused differential decreases in the relaxations induced by acetylcholine and nitroprusside. Only PAR2-HD lowered systolic arterial pressures in WT, when compared with all of the other groups. TNF-α and IL-1β plasma concentrations were not different among the groups. We conclude that the systolic blood pressure of unrestrained mice can be lowered by chronic in-vivo activation of PAR2; however, this effect is countered by receptor desensitization and the concomitant development of endothelium and vascular dysfunction.
Collapse
Affiliation(s)
- Keon H. Hughes
- Cardiovascular Research Group, Division of BioMedical Sciences, Memorial University, 300 Prince Philip Drive, St. John's, NL A1B 3V6, Canada
| | - Enoka P. Wijekoon
- Cardiovascular Research Group, Division of BioMedical Sciences, Memorial University, 300 Prince Philip Drive, St. John's, NL A1B 3V6, Canada
| | - James E. Valcour
- Division of Community Health and Humanities, Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St. John's, NL A1B 3V6, Canada
| | - Elizabeth W. Chia
- Cardiovascular Research Group, Division of BioMedical Sciences, Memorial University, 300 Prince Philip Drive, St. John's, NL A1B 3V6, Canada
| | - John J. McGuire
- Cardiovascular Research Group, Division of BioMedical Sciences, Memorial University, 300 Prince Philip Drive, St. John's, NL A1B 3V6, Canada
| |
Collapse
|
13
|
Lucking AJ, Gibson KR, Paterson EE, Faratian D, Ludlam CA, Boon NA, Fox KAA, Newby DE. Endogenous tissue plasminogen activator enhances fibrinolysis and limits thrombus formation in a clinical model of thrombosis. Arterioscler Thromb Vasc Biol 2013; 33:1105-11. [PMID: 23471234 DOI: 10.1161/atvbaha.112.300395] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Using a clinical model of deep arterial injury, we assessed the ability of exogenous and endogenous tissue plasminogen activator (t-PA) to limit acute in situ thrombus formation. APPROACH AND RESULTS Ex vivo thrombus formation was assessed in the Badimon chamber at low and high shear rates in 2 double-blind randomized cross-over studies of 20 healthy volunteers during extracorporeal administration of recombinant t-PA (0, 40, 200, and 1000 ng/mL) or during endogenous t-PA release stimulated by intra-arterial bradykinin infusion in the presence or absence of oral enalapril. Recombinant t-PA caused a dose-dependent reduction in thrombus area under low and high shear conditions (P<0.001 for all). Intra-arterial bradykinin increased plasma t-PA concentrations in the chamber effluent (P<0.01 for all versus saline) that was quadrupled in the presence of enalapril (P<0.0001 versus placebo). These increases were accompanied by an increase in plasma D-dimer concentration (P<0.005 for all versus saline) and, in the presence of enalapril, a reduction in thrombus area in the low shear (16±5; P=0.03) and a trend toward a reduction in the high shear chamber (13±7%; P=0.07). CONCLUSIONS Using a well-characterized clinical model of coronary arterial injury, we demonstrate that endogenous t-PA released from the vascular endothelium enhances fibrinolysis and limits in situ thrombus propagation. These data support a crucial role for the endogenous fibrinolytic system in vivo and suggest that continued exploration and manipulation of its therapeutic potential are warranted.
Collapse
Affiliation(s)
- Andrew J Lucking
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4SU, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Venkatasubramanian S, Griffiths ME, McLean SG, Miller MR, Luo R, Lang NN, Newby DE. Vascular effects of urocortins 2 and 3 in healthy volunteers. J Am Heart Assoc 2013; 2:e004267. [PMID: 23525432 PMCID: PMC3603262 DOI: 10.1161/jaha.112.004267] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Urocortin 2 and urocortin 3 are endogenous peptides with an emerging role in cardiovascular pathophysiology. We assessed their pharmacodynamic profile and examined the role of the endothelium in mediating their vasomotor effects in vivo in man. METHODS AND RESULTS Eighteen healthy male volunteers (23±4 years) were recruited into a series of double-blind, randomized crossover studies using bilateral forearm venous occlusion plethysmography during intra-arterial urocortin 2 (3.6 to 120 pmol/min), urocortin 3 (1.2 to 36 nmol/min), and substance P (2 to 8 pmol/min) in the presence or absence of inhibitors of cyclooxygenase (aspirin), cytochrome P450 metabolites of arachidonic acid (fluconazole), and nitric oxide synthase (L-NMMA). Urocortins 2 and 3 evoked arterial vasodilatation (P<0.0001) without tachyphylaxis but with a slow onset and offset of action. Inhibition of nitric oxide synthase with L-NMMA reduced vasodilatation to substance P and urocortin 2 (P≤0.001 for both) but had little effect on urocortin 3 (P>0.05). Neither aspirin nor fluconazole affected vasodilatation induced by any of the infusions (P>0.05 for all). In the presence of all 3 inhibitors, urocortin 2- and urocortin 3-induced vasodilatation was attenuated (P<0.001 for all) to a greater extent than with L-NMMA alone (P≤0.005). CONCLUSIONS Urocortins 2 and 3 cause potent and prolonged arterial vasodilatation without tachyphylaxis. These vasomotor responses are at least partly mediated by endothelial nitric oxide and cytochrome P450 metabolites of arachidonic acid. The role of urocortins 2 and 3 remains to be explored in the setting of human heart failure, but they have the potential to have major therapeutic benefits. CLINICAL TRIAL REGISTRATION http://www.clinicaltrials.gov//. Unique identifier: NCT01096706 and NCT01296607.
Collapse
|
15
|
Chang CJ, Chen YC, Kao YH, Lin YK, Chen SA, Chen YJ. Dabigatran and Thrombin Modulate Electrophysiological Characteristics of Pulmonary Vein and Left Atrium. Circ Arrhythm Electrophysiol 2012; 5:1176-83. [DOI: 10.1161/circep.112.971556] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Chien-Jung Chang
- From the Graduate Institute of Clinical Medicine, College of Medicine (C-J.C., Y-K.L., Y-J.C.), Department of Medical Education and Research, Wan Fang Hospital (Y-H.K.), and Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital (Y-K.L., Y-J.C.), Taipei Medical University, Taipei, Taiwan; Division of Cardiology, Tungs’ Taichung Metroharbour Hospital, Taichung, Taiwan (C-J.C.); Department of Biomedical Engineering and Institute of Physiology, National Defense Medical
| | - Yao-Chang Chen
- From the Graduate Institute of Clinical Medicine, College of Medicine (C-J.C., Y-K.L., Y-J.C.), Department of Medical Education and Research, Wan Fang Hospital (Y-H.K.), and Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital (Y-K.L., Y-J.C.), Taipei Medical University, Taipei, Taiwan; Division of Cardiology, Tungs’ Taichung Metroharbour Hospital, Taichung, Taiwan (C-J.C.); Department of Biomedical Engineering and Institute of Physiology, National Defense Medical
| | - Yu-Hsun Kao
- From the Graduate Institute of Clinical Medicine, College of Medicine (C-J.C., Y-K.L., Y-J.C.), Department of Medical Education and Research, Wan Fang Hospital (Y-H.K.), and Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital (Y-K.L., Y-J.C.), Taipei Medical University, Taipei, Taiwan; Division of Cardiology, Tungs’ Taichung Metroharbour Hospital, Taichung, Taiwan (C-J.C.); Department of Biomedical Engineering and Institute of Physiology, National Defense Medical
| | - Yung-Kuo Lin
- From the Graduate Institute of Clinical Medicine, College of Medicine (C-J.C., Y-K.L., Y-J.C.), Department of Medical Education and Research, Wan Fang Hospital (Y-H.K.), and Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital (Y-K.L., Y-J.C.), Taipei Medical University, Taipei, Taiwan; Division of Cardiology, Tungs’ Taichung Metroharbour Hospital, Taichung, Taiwan (C-J.C.); Department of Biomedical Engineering and Institute of Physiology, National Defense Medical
| | - Shih-Ann Chen
- From the Graduate Institute of Clinical Medicine, College of Medicine (C-J.C., Y-K.L., Y-J.C.), Department of Medical Education and Research, Wan Fang Hospital (Y-H.K.), and Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital (Y-K.L., Y-J.C.), Taipei Medical University, Taipei, Taiwan; Division of Cardiology, Tungs’ Taichung Metroharbour Hospital, Taichung, Taiwan (C-J.C.); Department of Biomedical Engineering and Institute of Physiology, National Defense Medical
| | - Yi-Jen Chen
- From the Graduate Institute of Clinical Medicine, College of Medicine (C-J.C., Y-K.L., Y-J.C.), Department of Medical Education and Research, Wan Fang Hospital (Y-H.K.), and Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital (Y-K.L., Y-J.C.), Taipei Medical University, Taipei, Taiwan; Division of Cardiology, Tungs’ Taichung Metroharbour Hospital, Taichung, Taiwan (C-J.C.); Department of Biomedical Engineering and Institute of Physiology, National Defense Medical
| |
Collapse
|
16
|
Palayoor ST, J-Aryankalayil M, Makinde AY, Cerna D, Falduto MT, Magnuson SR, Coleman CN. Gene expression profile of coronary artery cells treated with nonsteroidal anti-inflammatory drugs reveals off-target effects. J Cardiovasc Pharmacol 2012; 59:487-99. [PMID: 22668799 PMCID: PMC3370396 DOI: 10.1097/fjc.0b013e31824ba6b5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) have come under scrutiny because of the gastrointestinal, renal, and cardiovascular toxicity associated with prolonged use of these drugs. The purpose of this study was to identify molecular targets for NSAIDs related to cellular toxicity with a view to optimize drug efficacy in the clinic. Coronary artery smooth muscle cells and endothelial cells were treated with low (clinically achievable) and high (typically used in preclinical studies) concentrations of celecoxib, NS398, and ibuprofen for 24 hours. NSAIDs-induced gene expression changes were evaluated by microarray analysis and validated by real-time reverse-transcription polymerase chain reaction and western blotting. The functional significance of differentially expressed genes was evaluated by Ingenuity Pathway Analysis. At high concentrations, NSAIDs altered the expression of genes regulating cell proliferation and cell death. NSAIDs also altered genes associated with cardiovascular functions including inflammation, thrombosis, fibrinolysis, coronary artery disease, and hypertension. The gene expression was most impacted by ibuprofen, celecoxib, and NS398, in that order. This study revealed that NSAIDs altered expression of an array of genes associated with cardiovascular events and emphasizes the potential for fingerprinting drugs in preclinical studies to assess the potential drug toxicity and to optimize the drug efficacy in clinical settings.
Collapse
MESH Headings
- Anti-Inflammatory Agents, Non-Steroidal/administration & dosage
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Blotting, Western
- Celecoxib
- Cell Proliferation/drug effects
- Cells, Cultured
- Coronary Vessels/cytology
- Coronary Vessels/drug effects
- Coronary Vessels/metabolism
- Dose-Response Relationship, Drug
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Gene Expression Profiling
- Gene Expression Regulation/drug effects
- Humans
- Ibuprofen/pharmacology
- Microarray Analysis
- Molecular Targeted Therapy
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Nitrobenzenes/administration & dosage
- Nitrobenzenes/pharmacology
- Pyrazoles/administration & dosage
- Pyrazoles/pharmacology
- Reverse Transcriptase Polymerase Chain Reaction
- Sulfonamides/administration & dosage
- Sulfonamides/pharmacology
Collapse
Affiliation(s)
- Sanjeewani T Palayoor
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Building # 10, Room B3B406, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
17
|
Lee H, Hamilton JR. Physiology, pharmacology, and therapeutic potential of protease-activated receptors in vascular disease. Pharmacol Ther 2012; 134:246-59. [DOI: 10.1016/j.pharmthera.2012.01.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 01/17/2012] [Indexed: 01/09/2023]
|
18
|
Abstract
Vascular endothelium is a key regulator of homeostasis. In physiological conditions it mediates vascular dilatation, prevents platelet adhesion, and inhibits thrombin generation. However, endothelial dysfunction caused by physical injury of the vascular wall, for example during balloon angioplasty, acute or chronic inflammation, such as in atherothrombosis, creates a proinflammatory environment which supports leukocyte transmigration toward inflammatory sites. At the same time, the dysfunction promotes thrombin generation, fibrin deposition, and coagulation. The serine protease thrombin plays a pivotal role in the coagulation cascade. However, thrombin is not only the key effector of coagulation cascade; it also plays a significant role in inflammatory diseases. It shows an array of effects on endothelial cells, vascular smooth muscle cells, monocytes, and platelets, all of which participate in the vascular pathophysiology such as atherothrombosis. Therefore, thrombin can be considered as an important modulatory molecule of vascular homeostasis. This review summarizes the existing evidence on the role of thrombin in vascular inflammation.
Collapse
|
19
|
Daga S, Shepherd JG, Callaghan JGS, Hung RK, Dawson DK, Padfield GJ, Hey SY, Cartwright RA, Newby DE, Fitzgerald JR. Platelet receptor polymorphisms do not influence Staphylococcus aureus-platelet interactions or infective endocarditis. Microbes Infect 2011; 13:216-25. [PMID: 21044892 PMCID: PMC3036801 DOI: 10.1016/j.micinf.2010.10.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 10/19/2010] [Accepted: 10/20/2010] [Indexed: 12/01/2022]
Abstract
Cardiac vegetations result from bacterium-platelet adherence, activation and aggregation, and are associated with increased morbidity and mortality in infective endocarditis. The GPIIb/IIIa and FcγRIIa platelet receptors play a central role in platelet adhesion, activation and aggregation induced by endocarditis pathogens such as Staphylococcus aureus, but the influence of known polymorphisms of these receptors on the pathogenesis of infective endocarditis is unknown. We determined the GPIIIa platelet antigen Pl(A1/A2) and FcγRIIa H131R genotype of healthy volunteers (n = 160) and patients with infective endocarditis (n = 40), and investigated the influence of these polymorphisms on clinical outcome in infective endocarditis and S. aureus-platelet interactions in vitro. Platelet receptor genotype did not correlate with development of infective endocarditis, vegetation characteristics on echocardiogram or the composite clinical end-point of embolism, heart failure, need for surgery or mortality (P > 0.05 for all), even though patients with the GPIIIa Pl(A1/A1) genotype had increased in vivo platelet activation (P = 0.001). Furthermore, neither GPIIIa Pl(A1/A2) nor FcγRIIa H131R genotype influenced S. aureus-induced platelet adhesion, activation or aggregation in vitro (P > 0.05). Taken together, our data suggest that the GPIIIa and FcγRIIa platelet receptor polymorphisms do not influence S. aureus-platelet interactions in vitro or the clinical course of infective endocarditis.
Collapse
Affiliation(s)
- Shruti Daga
- Center for Infectious Diseases and The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, United Kingdom
- Center for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, United Kingdom
| | - James G. Shepherd
- Center for Infectious Diseases and The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, United Kingdom
- Center for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, United Kingdom
| | - J. Garreth S. Callaghan
- Center for Infectious Diseases and The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, United Kingdom
- Center for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, United Kingdom
| | - Rachel K.Y. Hung
- Center for Infectious Diseases and The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, United Kingdom
- Center for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, United Kingdom
| | - Dana K. Dawson
- Center for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, United Kingdom
| | - Gareth J. Padfield
- Center for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, United Kingdom
| | - Shi Y. Hey
- Center for Infectious Diseases and The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, United Kingdom
- Center for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, United Kingdom
| | - Robyn A. Cartwright
- Center for Infectious Diseases and The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, United Kingdom
| | - David E. Newby
- Center for Cardiovascular Science, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, United Kingdom
| | - J. Ross Fitzgerald
- Center for Infectious Diseases and The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Chancellor’s Building, Edinburgh EH16 4SB, United Kingdom
| |
Collapse
|
20
|
Abstract
Despite major advances in antiplatelet therapies, recurrent cardiovascular events remain high after acute coronary syndrome. Furthermore, incremental benefits achieved in the reduction of atherothrombotic events have almost always been at the expense of hemorrhagic side effects. Thrombin is the most potent platelet activating factor known and it makes important interactions with the endothelium and vascular smooth muscle with proinflammatory, proatherogenic effects. Distinct from its activity within the coagulation cascade, thrombin mediates these effects via protease-activated receptor type 1 (PAR-1) in man. This review discusses the role of PAR-1 in the vasculature and the development of novel PAR-1 antagonists. These drugs may provide important antiatherothrombotic effects without attendant bleeding complications and could represent a major breakthrough for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Ninian N Lang
- Centre for Cardiovascular Science, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, UK
| | | | | |
Collapse
|
21
|
Chaudhry R, Chaudhry FA, Huynh T, Lader E, Rashid S, Okrainec K, Wou K, Eisenberg MJ. Effect of smoking on age at the time of coronary artery bypass graft surgery; baseline data results from the ROSETTA-CABG registry. HEART ASIA 2010; 2:48-51. [PMID: 27325942 DOI: 10.1136/ha.2009.001586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/20/2010] [Indexed: 11/04/2022]
Abstract
BACKGROUND Coronary artery disease (CAD) is a leading cause of death. The aetiology of this disease is not known, but many important risk factors have been recognised. OBJECTIVE To evaluate the effect of smoking on age at the time of coronary artery bypass graft surgery (CABG), and to examine this finding in the light of medical literature. METHODS The authors recruited patients immediately after CABG in a prospective, study in 16 centres and enrolled 408 patients, of which 395 were ultimately analysed. RESULTS Among the 395 patients analysed, there were 60 smokers and 335 non-smokers. The smokers were 8.4 years younger than non-smokers at the time of index CABG. The average age of smokers was 55.79.0 years, and that of non-smokers was 64.1±9.9 years (p<0.001). Hyperlipidaemia was present in 76.7% of smokers and 74.6% of non-smokers (p-NS). Hypertension was present in 58.3% of smokers and 63.9% of non-smokers (p-NS). Diabetes mellitus was present in 21.3% of smokers and 29.3% of non smokers (p-NS). Left ventricular ejection fraction was 53.0±10.5% in smokers and 53.3 ±13.8% in non-smokers (p-NS). Myocardial infarction had occurred in 41.7% of smokers and 35.5% of non-smokers (p-NS). CONCLUSION Smoking accelerates atherosclerosis and coronary thrombosis resulting in severe form of CAD that cannot be managed by medications or PCI, and requires coronary artery bypass graft surgery (CABG) 8.4 years earlier than non-smokers.
Collapse
Affiliation(s)
| | - Fahd A Chaudhry
- University Physician Health Care, University of Arizona, Tucson, Arizona, USA
| | - Thao Huynh
- Division of Cardiology, Montreal General Hospital, Montreal, Quebec, Canada
| | - Ellis Lader
- Mid Valley Cardiology, Kingston, New York, USA
| | - Saira Rashid
- King Edward Medical University, Lahore, Pakistan
| | - Karen Okrainec
- Division of Internal Medicine, McGill University Health Center, Montreal, Quebec, Canada
| | - Karen Wou
- Division of Internal Medicine, McGill University Health Center, Montreal, Quebec, Canada
| | - Mark J Eisenberg
- Divisions of Cardiology and Clinical Epidemiology, Jewish General Hospital, Montreal, Quebec, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
22
|
Protease-activated receptors, cyclo-oxygenases and pro-angiogenic signalling in endothelial cells. Biochem Soc Trans 2009; 37:1179-83. [DOI: 10.1042/bst0371179] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
COX (cyclo-oxygenase)-2 and members of the PAR (protease-activated receptor) family (PARs 1–4) are highly overexpressed in a number of angiogenesis-dependent pathologies, including advanced atherosclerosis and cancer. An appreciation of the potential role(s) of PARs and COX enzymes in physiological angiogenesis is, however, currently lacking. Exposure of human endothelial cells to serine proteases (e.g. thrombin) or to PAR-selective agonist peptides leads to a wide range of cellular responses, including enhanced expression of COX-2, and we have shown that this induction depends on activation of classic pro-inflammatory signalling elements [e.g. MAPKs (mitogen-activated protein kinases) and NF-κB (nuclear factor κB)]. Our current studies suggest that COX-2-derived mediators are important autocrine regulators of PAR-stimulated angiogenesis. This mechanism could help us to explain how this novel family of receptors couple vascular inflammation with repair and angiogenesis in health and disease.
Collapse
|
23
|
Chelliah R, Lucking AJ, Tattersall L, Daga S, Beresford-Cleary NJ, Cortas K, Fox KAA, Feuerstein GZ, Connolly TM, Newby DE. P-selectin antagonism reduces thrombus formation in humans. J Thromb Haemost 2009; 7:1915-9. [PMID: 19691482 DOI: 10.1111/j.1538-7836.2009.03587.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Interaction of P-selectin with its glycoprotein ligand (P-selectin glycoprotein ligand type 1) mediates inflammatory processes that may also include vascular thrombosis. Platelet P-selectin expression is increased in patients with coronary heart disease, and its antagonism represents a potential future therapeutic target for the prevention and treatment of atherothrombosis. AIM To investigate the effects of the novel small molecule P-selectin antagonist PSI-697 on thrombus formation in humans. METHODS AND RESULTS In a double-blind randomized crossover study, thrombus formation was measured in 12 healthy volunteers, using the Badimon ex vivo perfusion chamber under conditions of low and high shear stress. Saline placebo, low-dose (2 m) and high-dose (20 m) PSI-697 and the glycoprotein IIb-IIIa receptor antagonist tirofiban (50 ng mL(-1)) were administered into the extracorporeal circuit prior to the perfusion chamber. As compared with saline placebo, blockade of platelet glycoprotein IIb-IIIa receptor with tirofiban produced 28% and 56% reductions in thrombus formation in the low-shear and high-shear chambers, respectively. PSI-697 caused a dose-dependent, but more modest, reduction in thrombus formation. Low-dose PSI-796 (2 m) reduced total thrombus area by 14% (P = 0.04) and 30% (P = 0.0002) in the low-shear and high-shear chambers, respectively. At the high dose (20 m), PSI-697 reduced total thrombus area by 18% (P = 0.0094) and 41% (P = 0.0008) in the low-shear and high-shear chambers, respectively. CONCLUSIONS P-selectin antagonism with PSI-697 reduces ex vivo thrombus formation in humans. These findings provide further evidence that P-selectin antagonism may be a potential target for the prevention and treatment of cardiovascular disease.
Collapse
Affiliation(s)
- R Chelliah
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Marked impairment of protease-activated receptor type 1-mediated vasodilation and fibrinolysis in cigarette smokers: smoking, thrombin, and vascular responses in vivo. J Am Coll Cardiol 2008; 52:33-9. [PMID: 18582632 DOI: 10.1016/j.jacc.2008.04.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 03/25/2008] [Accepted: 04/02/2008] [Indexed: 11/23/2022]
Abstract
OBJECTIVES We sought to test the hypothesis that cigarette smoking adversely alters protease-activated receptor type 1 (PAR-1)-mediated vascular effects in vivo in humans. BACKGROUND Distinct from its role in the coagulation cascade, thrombin exerts its major cellular and cardiovascular actions via PAR-1. The activation of PAR-1 causes endothelium-dependent arterial vasodilation and the release of endogenous fibrinolytic factors. METHODS Forearm blood flow was measured with venous occlusion plethysmography in 12 cigarette smokers and 12 age- and gender-matched nonsmokers during intrabrachial infusions of PAR-1-activating-peptide (SFLLRN; 5 to 50 nmol/min), bradykinin (100 to 1,000 pmol/min), and sodium nitroprusside (2 to 8 mug/min). Plasma tissue plasminogen activator (t-PA) and plasminogen-activator inhibitor 1 antigen and activity concentrations were measured throughout the experiment. RESULTS All agonists caused dose-dependent increases in forearm blood flow (p < 0.0001 for all). Although bradykinin and sodium nitroprusside caused similar vasodilation, SFLLRN-induced vasodilation was attenuated in smokers (p = 0.04). Smokers had modest reductions in bradykinin-induced active t-PA release (reduced by 37%, p = 0.03) and had a marked impairment of SFLLRN-induced t-PA antigen (p = 0.02) and activity (p = 0.006) release, with a 96% reduction in overall net t-PA antigen release. The use of SFLLRN also caused similar (p = NS) increases in inactive plasminogen-activator inhibitor 1 in both smokers and nonsmokers (p <or= 0.002 for both). CONCLUSIONS Cigarette smoking causes marked impairment of PAR-1-mediated endothelial vasomotor and fibrinolytic function. Relative arterial stasis and near abolition of t-PA release will strongly promote clot propagation and vessel occlusion. These findings suggest a major contribution of impaired endothelial PAR-1 action to the increased atherothrombotic risk of cigarette smokers.
Collapse
|
25
|
Gudmundsdóttir IJ, Lang NN, Boon NA, Ludlam CA, Webb DJ, Fox KA, Newby DE. Role of the endothelium in the vascular effects of the thrombin receptor (protease-activated receptor type 1) in humans. J Am Coll Cardiol 2008; 51:1749-56. [PMID: 18452780 DOI: 10.1016/j.jacc.2007.12.047] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2007] [Revised: 11/21/2007] [Accepted: 12/17/2007] [Indexed: 11/16/2022]
Abstract
OBJECTIVES The purpose of this study was to determine the role of the endothelium in the vascular actions of protease-activated receptor type 1 (PAR-1) activation in vivo in man. BACKGROUND Thrombin is central to the pathophysiology of atherothrombosis. Its cellular actions are mediated via PAR-1. Protease-activated receptor type 1 activation causes arterial vasodilation, venoconstriction, platelet activation, and tissue-type plasminogen activator release in man. METHODS Dorsal hand vein diameter was measured in 6 healthy volunteers before and after endothelial denudation. Forearm arterial blood flow, plasma fibrinolytic factors, and platelet activation were measured in 24 healthy volunteers during venous occlusion plethysmography. The effects of inhibition of prostacyclin, nitric oxide (NO), and endothelium-derived hyperpolarizing factor on PAR-1 responses were assessed during coadministration of aspirin, the "NO clamp" (L-N(G)-monomethyl arginine and sodium nitroprusside), and tetraethylammonium ion, respectively. RESULTS Endothelial denudation did not affect PAR-1-evoked venoconstriction (SFLLRN; 0.05 to 15 nmol/min). Although aspirin had no effect, SFLLRN-induced vasodilation (5 to 50 nmol/min) was attenuated by the NO clamp (p < 0.0001) and tetraethylammonium ion (p < 0.05) and abolished by their combination (p < 0.01). The NO clamp augmented SFLLRN-induced tissue-type plasminogen activator and plasminogen activator inhibitor type 1 antigen (p < 0.0001) release, but tetraethylammonium ion and aspirin had no effect. SFLLRN-induced platelet activation was unaffected by NO or prostacyclin inhibition. CONCLUSIONS Acting via PAR-1, thrombin causes contrasting effects in the human vasculature and has a major interaction with the endothelium. This highlights the critical importance of endothelial function during acute arterial injury and intravascular thrombosis, as occurs in cardiovascular events including myocardial infarction and stroke.
Collapse
|
26
|
Cleator JH, Vaughan DE. Clinical Implications of the Contrasting Effects of In Vivo Thrombin Receptor Activation (Protease-Activated Receptor Type 1) on the Human Vasculature⁎⁎Editorials published in the Journal of the American College of Cardiology reflect the views of the authors and do not necessarily represent the views of JACC or the American College of Cardiology. J Am Coll Cardiol 2008; 51:1757-9. [DOI: 10.1016/j.jacc.2008.01.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2007] [Revised: 01/09/2008] [Accepted: 01/15/2008] [Indexed: 01/03/2023]
|
27
|
McBane RD, Leadley RJ, Baxi SM, Karnicki K, Wysokinski W. Iliac venous stenting: antithrombotic efficacy of PD0348292, an oral direct Factor Xa inhibitor, compared with antiplatelet agents in pigs. Arterioscler Thromb Vasc Biol 2007; 28:413-8. [PMID: 18096830 DOI: 10.1161/atvbaha.107.158691] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The clinical use of venous stents is increasing dramatically. Although antiplatelet agents are required for arterial stent patency, optimal thrombo-prophylaxis after venous stenting remains undefined. To address this issue, PD0348292, a direct Factor Xa inhibitor, was compared with antiplatelet therapy in a porcine venous stent model. METHODS AND RESULTS Four hours before stent deployment, pigs (n=5 to 6 per group) received oral PD0348292 at 0.4, 0.9, 4.3 mg/kg, or 0.4 mg/kg plus aspirin (325 mg). Aspirin, clopidogrel (75 mg), aspirin plus clopidogrel, or vehicle (n=10) were administered daily for 2 days before the procedure. Two hours after stent placement, thrombi were quantified by autologous (111)In-platelet content and weights. Thrombus weight and platelet deposition were significantly reduced by PD0348292 at 0.4 (49+/-79 mg and 110+/-145x10(6)/cm2), 0.9 (5+/-6 mg and 107+/-128x10(6)/cm2), 4.3 mg/kg (0+/-0 mg and 87+/-125x10(6)/cm2), and PD348292 plus aspirin (20+/-40 mg and 157+/-70x10(6)/cm2) compared with vehicle (402+/-226 mg; 584+/-454x10(6)/cm2). Despite prolonging bleeding times and inhibiting platelet aggregation, neither aspirin (567+/-683 mg and 533+/-622x10(6)/cm2), clopidogrel (404+/-349 mg and 178+/-101x10(6)/cm2), nor aspirin plus clopidogrel (247+/-261 mg and 231+/-266x10(6)/cm2) significantly decreased stent thrombosis. CONCLUSIONS PD0348292 completely inhibited thrombosis after venous stenting. Platelet accretion in these venous thrombi appear to involve pathways distinct from arachidonate metabolism or ADP P2Y12 receptor activation.
Collapse
Affiliation(s)
- Robert D McBane
- Section of Hematology Research, Mayo Clinic, 200 First Street, Rochester, MN 55905, USA.
| | | | | | | | | |
Collapse
|
28
|
Bae JS, Yang L, Manithody C, Rezaie AR. The ligand occupancy of endothelial protein C receptor switches the protease-activated receptor 1-dependent signaling specificity of thrombin from a permeability-enhancing to a barrier-protective response in endothelial cells. Blood 2007; 110:3909-16. [PMID: 17823308 PMCID: PMC2190610 DOI: 10.1182/blood-2007-06-096651] [Citation(s) in RCA: 190] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Recent studies have indicated that activated protein C (APC) may exert its cytoprotective and anti-inflammatory activities through the endothelial protein C receptor (EPCR)-dependent cleavage of protease-activated receptor 1 (PAR-1) on vascular endothelial cells. Noting that (1) the activation of protein C on endothelial cells requires thrombin, (2) relative to APC, thrombin cleaves PAR-1 with approximately 3 to 4 orders of magnitude higher catalytic efficiency, and (3) PAR-1 is a target for the proinflammatory activity of thrombin, it is not understood how APC can elicit a protective signaling response through the cleavage of PAR-1 when thrombin is present. In this study, we demonstrate that EPCR is associated with caveolin-1 in lipid rafts of endothelial cells and that its occupancy by the gamma-carboxyglutamic acid (Gla) domain of protein C/APC leads to its dissociation from caveolin-1 and recruitment of PAR-1 to a protective signaling pathway through coupling of PAR-1 to the pertussis toxin-sensitive G(i)-protein. Thus, when EPCR is bound by protein C, the PAR-1 cleavage-dependent protective signaling responses in endothelial cells can be mediated by either thrombin or APC. These results provide a new paradigm for understanding how PAR-1 and EPCR participate in protective signaling events in endothelial cells.
Collapse
Affiliation(s)
- Jong-Sup Bae
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, MO 63104, USA
| | | | | | | |
Collapse
|
29
|
Fujiyoshi T, Hirano K, Hirano M, Nishimura J, Takahashi S, Kanaide H. Plasmin induces endothelium-dependent nitric oxide-mediated relaxation in the porcine coronary artery. Arterioscler Thromb Vasc Biol 2007; 27:949-54. [PMID: 17272753 DOI: 10.1161/01.atv.0000259360.33203.00] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Plasmin is a key enzyme in fibrinolysis. We attempted to determine the possible role of plasmin in the regulation of vascular tone, while also investigating the mechanism of plasmin-induced vasorelaxation. METHODS AND RESULTS In porcine coronary artery, plasmin induced an endothelium-dependent relaxation. This relaxing effect was mostly abolished by a proteinase inhibitor, a plasmin inhibitor, or a nitric oxide (NO) synthase inhibitor. The preceding stimulation with plasmin significantly inhibited the subsequent relaxation induced by thrombin but not that induced by proteinase-activated receptor-1-activating peptide. The relaxation induced by trypsin and substance P remained unaffected by the preceding plasmin stimulation. The pretreatment with plasmin, thrombin, or trypsin significantly attenuated the plasmin-induced relaxation. In porcine coronary artery endothelial cells (PCAECs) and human umbilical vein endothelial cells (HUVECs), plasmin induced a transient elevation in the cytosolic Ca2+ concentrations ([Ca2+]i). The preceding stimulation with plasmin inhibited the subsequent [Ca2+]i elevation induced by thrombin but not that induced by trypsin. In PCAECs, plasmin concentration-dependently induced NO production. CONCLUSIONS The present study demonstrated, for the first time, that plasmin induced an endothelium-dependent NO-mediated relaxation in the porcine coronary artery, while also showing plasmin to specifically inactivate the thrombin receptor.
Collapse
Affiliation(s)
- Tetsuhiro Fujiyoshi
- Division of Molecular Cardiology, Research Institute of Angiocardiology, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | |
Collapse
|