1
|
Han X, Nieman MT. The domino effect triggered by the tethered ligand of the protease activated receptors. Thromb Res 2020; 196:87-98. [PMID: 32853981 DOI: 10.1016/j.thromres.2020.08.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/23/2020] [Accepted: 08/03/2020] [Indexed: 12/20/2022]
Abstract
Protease activated receptors (PARs) are G-protein coupled receptors (GPCRs) that have a unique activation mechanism. Unlike other GPCRs that can be activated by free ligands, under physiological conditions, PARs are activated by the tethered ligand, which is a part of their N-terminus that is unmasked by proteolysis. It has been 30 years since the first member of the family, PAR1, was identified. In this review, we will discuss this unique tethered ligand mediate receptor activation of PARs in detail: how they interact with the proteases, the complex structural rearrangement of the receptors upon activation, and the termination of the signaling. We also summarize the structural studies of the PARs and how single nucleotide polymorphisms impact the receptor reactivity. Finally, we review the current strategies for inhibiting PAR function with therapeutic targets for anti-thrombosis. The focus of this review is PAR1 and PAR4 as they are the thrombin signal mediators on human platelets and therapeutics targets. We also include the structural studies of PAR2 as it informs the mechanism of action for PARs in general.
Collapse
Affiliation(s)
- Xu Han
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Marvin T Nieman
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
2
|
Hamilton JR, Trejo J. Challenges and Opportunities in Protease-Activated Receptor Drug Development. Annu Rev Pharmacol Toxicol 2016; 57:349-373. [PMID: 27618736 DOI: 10.1146/annurev-pharmtox-011613-140016] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Protease-activated receptors (PARs) are a unique class of G protein-coupled receptors (GPCRs) that transduce cellular responses to extracellular proteases. PARs have important functions in the vasculature, inflammation, and cancer and are important drug targets. A unique feature of PARs is their irreversible proteolytic mechanism of activation that results in the generation of a tethered ligand that cannot diffuse away. Despite the fact that GPCRs have proved to be the most successful class of druggable targets, the development of agents that target PARs specifically has been challenging. As a consequence, researchers have taken a remarkable diversity of approaches to develop pharmacological entities that modulate PAR function. Here, we present an overview of the diversity of therapeutic agents that have been developed against PARs. We further discuss PAR biased signaling and the influence of receptor compartmentalization, posttranslational modifications, and dimerization, which are important considerations for drug development.
Collapse
Affiliation(s)
- Justin R Hamilton
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia
| | - JoAnn Trejo
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California 92093;
| |
Collapse
|
3
|
Wickham LA, Sitko G, Stranieri-Michener M, Handt L, Basso A, Fried S, Chu L, Maderia M, Owens K, Castriota G, Chen Z, Metzger JM, Imbriglio J, Wang X, Cai TQ. Differential anti-thrombotic benefit and bleeding risk profiles of antagonists of protease-activated receptor 1 and 4 in Cynomolgus Macaques. Thromb Res 2016; 145:133-9. [PMID: 27318768 DOI: 10.1016/j.thromres.2016.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/24/2016] [Accepted: 06/08/2016] [Indexed: 11/30/2022]
Abstract
Platelet activation plays a crucial role in hemostasis and thrombosis. Thrombin, the most potent stimulus of platelet activation, mediates platelet activation via the protease activated receptors (PARs). The platelet PAR repertoire in mediating thrombin's action differs across species. Only nonhuman primate (NHP) platelet activation is known to be similar to humans, mediated by PAR1 and PAR4, hence limiting translational in vivo studies of PAR's role in thrombosis and hemostasis to NHPs. Earlier studies have demonstrated a range of distinct in vitro activities of PAR1 and 4 in platelet activation yet the implications of these events in vivo is unclear. The objective of this study is to investigate and compare the roles of PAR1 and PAR4 in hemostasis and thrombosis in a relevant animal species. NHP models for pharmacokinetic, ex vivo platelet aggregation responses, FeCI3 injury-mediated arterial thrombosis and template bleeding were developed in Cynomolgus Macaques. Potent and selective small molecule antagonists of PAR1 and PAR4 were characterized in an array of in vitro assays, and subsequently examined head-to-head in the NHP models. Treatment of NHPs with antagonists of PAR1 or PAR4 both resulted in strong inhibition of ex vivo platelet aggregation. At doses that led to similar inhibition of platelet aggregation, animals treated with the PAR4 antagonist showed similar levels of anti-thrombotic efficacy, but longer bleeding times, compared to animals treated with the PAR1 antagonist. These findings suggest that PAR1 antagonism will likely produce a larger therapeutic index (ie. a larger anti-thrombotic efficacy over bleeding risk margin) than PAR4 antagonism.
Collapse
Affiliation(s)
- L Alexandra Wickham
- Department of In Vivo Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Gary Sitko
- Department of Safety, Merck Research Laboratories, Kenilworth, NJ, USA
| | | | - Larry Handt
- Department of Safety, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Andrea Basso
- Department of In Vitro Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Steven Fried
- Department of In Vitro Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Lin Chu
- Department of Formulation, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Maria Maderia
- Department of Drug Metabolism, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Karen Owens
- Department of Drug Metabolism, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Gino Castriota
- Department of Cardiometabolic Diseases, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Zhu Chen
- Department of Cardiometabolic Diseases, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Joseph M Metzger
- Department of In Vivo Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Jason Imbriglio
- Department of Medicinal Chemistry, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Xinkang Wang
- Department of Cardiometabolic Diseases, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Tian-Quan Cai
- Department of In Vivo Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA.
| |
Collapse
|
4
|
Schaff M, Gachet C, Mangin PH. [Anti-platelets without a bleeding risk: novel targets and strategies]. Biol Aujourdhui 2016; 209:211-28. [PMID: 26820829 DOI: 10.1051/jbio/2015023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Indexed: 01/29/2023]
Abstract
Anti-platelet agents such as aspirin, clopidogrel and antagonists of integrin αIIbβ3 allowed to efficiently reduce morbidity and mortality associated with arterial thrombosis. A major limit of these drugs is that they increase the risk of bleeding. During the last few years, several innovative anti-thrombotic strategies with a potentially low bleeding risk were proposed. These approaches target the collagen receptor glycoprotein (GP) VI, the GPIb/von Willebrand factor axis, the thrombin receptor PAR-1, the activated form of integrin αIIbβ3 or the ADP receptor P2Y1. While an antagonist of PAR-1 was recently marketed, the clinical proofs of the efficiency and safety of the other agents remain to be established. This review evaluates these new anti-platelet approaches toward safer anti-thrombotic therapies.
Collapse
Affiliation(s)
- Mathieu Schaff
- Atherothrombosis and Vascular Biology Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Australie
| | - Christian Gachet
- UMR_S949, INSERM, Etablissement Français du Sang (EFS)-Alsace, Université de Strasbourg, Strasbourg, France
| | - Pierre Henri Mangin
- UMR_S949, INSERM, Etablissement Français du Sang (EFS)-Alsace, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
5
|
Amaral Imbeloni A, Canevese Rahal S, da Silva Filho E, Mendonça Faria B, Bandeira da Silva W, Guimarães Oliveira K, Aihara ME, Barros Monteiro FO. Effect of age and sex on bone markers in Chlorocebus aethiops raised in captivity. J Med Primatol 2015; 45:3-11. [PMID: 26555766 DOI: 10.1111/jmp.12201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2015] [Indexed: 12/01/2022]
Abstract
BACKGROUND The objective was to evaluate the procollagen type I N-propeptide (PINP), osteocalcin (OC), β-crosslaps (β-CTX), and parathyroid hormone (PTH) in relation to age and sex of Chlorocebus aethiops in captivity. METHODS Seventy-three monkeys were divided into four age groups: AG1 (juvenile); AG2 (young adult); AG3 (adult); and AG4 (senile). An electrochemiluminescence immunoassay with an Elecsys 2010 analyzer was used to determine the serum markers of bone. RESULTS AND CONCLUSIONS Sex did not influence the results of the markers. However, the variables PINP, OC, and β-CTX were negatively correlated with age (r = -0.643; r = -0.711; r = -0.488; P < 0.001, respectively), and PTH was correlated positively with age (r = 0.418, P < 0.001). The data obtained can be used as biomarkers of bone metabolism reference intervals in healthy C. aethiops in captivity.
Collapse
Affiliation(s)
- Aline Amaral Imbeloni
- Centro Nacional de Primatas, Secretaria de Vigilância em Saúde, Ministério da Saúde, Ananindeua, PA, Brazil.,Programa de Pós-graduação de Saúde e Produção Animal da Amazônia, Universidade Federal Rural da Amazônia, Belém, PA, Brazil
| | - Sheila Canevese Rahal
- Departamento de Cirurgia Veterinária e Anestesiologia da, Faculdade de Medicina Veterinária de Zootecnia, Universidade Estadual Paulista, Botucatu, SP, Brazil
| | - Ednaldo da Silva Filho
- Programa de Pós-graduação de Saúde e Produção Animal da Amazônia, Universidade Federal Rural da Amazônia, Belém, PA, Brazil
| | - Bianca Mendonça Faria
- Programa de Pós-graduação de Saúde e Produção Animal da Amazônia, Universidade Federal Rural da Amazônia, Belém, PA, Brazil
| | - Wellington Bandeira da Silva
- Centro Nacional de Primatas, Secretaria de Vigilância em Saúde, Ministério da Saúde, Ananindeua, PA, Brazil.,Programa de Pós-graduação de Saúde e Produção Animal da Amazônia, Universidade Federal Rural da Amazônia, Belém, PA, Brazil
| | - Karol Guimarães Oliveira
- Centro Nacional de Primatas, Secretaria de Vigilância em Saúde, Ministério da Saúde, Ananindeua, PA, Brazil
| | - Mika Ester Aihara
- Centro Nacional de Primatas, Secretaria de Vigilância em Saúde, Ministério da Saúde, Ananindeua, PA, Brazil
| | | |
Collapse
|
6
|
Diehl P, Bode C, Duerschmied D. Clinical potential of vorapaxar in cardiovascular risk reduction in patients with atherosclerosis. Ther Clin Risk Manag 2015; 11:1133-8. [PMID: 26346960 PMCID: PMC4529257 DOI: 10.2147/tcrm.s55469] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Vorapaxar (ZONTIVITY™, formerly known as SCH 530348) is a specific, orally active antagonist of the protease-activated receptor-1 (PAR-1) on platelets. It inhibits thrombin-induced platelet activation by binding to the ectodomain of PAR-1. After animal studies and Phase II studies showed that vorapaxar sufficiently inhibits platelet activation without significantly increasing bleeding complications, safety and efficacy of vorapaxar were assessed in two large multicenter trials in patients with coronary artery disease and atherosclerosis. The Thrombin-Receptor Antagonist for Clinical Event Reduction in Acute Coronary Syndromes (TRACER) trial investigated safety and efficacy of vorapaxar in patients with an acute coronary syndrome without ST-segment elevation. The Trial to Assess the Effects of Vorapaxar in Preventing Heart Attack and Stroke in Patients With Atherosclerosis-Thrombolysis In Myocardial Infarction 50 (TRA 2°P-TIMI 50) investigated atherothrombotic events in patients with stable atherosclerosis. Results of both studies suggested that vorapaxar given in addition to standard antiplatelet therapy can reduce atherothrombotic events, but increases the risk of mild and moderate bleeding complications. This review article summarizes the main results of TRACER and TRA 2°P-TIMI 50 and suggests patient cohorts that might benefit from treatment with vorapaxar in addition to standard antiplatelet therapy.
Collapse
Affiliation(s)
- Philipp Diehl
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Freiburg, Germany
| | - Christoph Bode
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Freiburg, Germany
| | - Daniel Duerschmied
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Freiburg, Germany
| |
Collapse
|
7
|
French SL, Arthur JF, Tran HA, Hamilton JR. Approval of the first protease-activated receptor antagonist: Rationale, development, significance, and considerations of a novel anti-platelet agent. Blood Rev 2014; 29:179-89. [PMID: 25467961 DOI: 10.1016/j.blre.2014.10.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 10/27/2014] [Indexed: 12/12/2022]
Abstract
Twenty-three years after the discovery of the first thrombin receptor, now known as protease-activated receptor 1 (PAR1), the first drug targeting this receptor is available for human use. The PAR1 inhibitor, vorapaxar (Zontivity, MSD), was recently approved by the FDA for use in the USA for the prevention of thrombotic cardiovascular events in patients with a history of myocardial infarction or peripheral artery disease. In this review, we detail the rationale, development, as well as the clinical significance and considerations of vorapaxar, the original PAR antagonist and the latest anti-platelet agent in the pharmaco-armoury against arterial thrombosis.
Collapse
Affiliation(s)
- Shauna L French
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia; Department of Clinical Haematology, Monash University, Melbourne, Victoria, Australia
| | - Jane F Arthur
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia; Department of Clinical Haematology, Monash University, Melbourne, Victoria, Australia
| | - Huyen A Tran
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia; Department of Clinical Haematology, Monash University, Melbourne, Victoria, Australia
| | - Justin R Hamilton
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia; Department of Clinical Haematology, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
8
|
MHC polymorphism in Caribbean African green monkeys. Immunogenetics 2014; 66:353-60. [PMID: 24676686 DOI: 10.1007/s00251-014-0770-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 03/14/2014] [Indexed: 11/27/2022]
Abstract
African green monkeys (AGM) are among the most widely used nonhuman primate models used in various fields of medical research. One species of AGM that originated from West Africa, Chlorocebus sabaeus, was introduced three centuries ago in the Caribbean islands. We present here a systematic study of the major histocompatibility complex (MHC) polymorphism of Caribbean AGM which is currently frequently used as an animal model. We studied 54 animals originated from Barbados (N=25) or Saint Kitts (N=29). The MHC polymorphism was characterized by means of 17 MHC microsatellites spread across MHC and DRB genotyping by DGGE sequencing. We defined nine frequent MHC haplotypes of which two were found in the two insular populations suggesting either past exchanges between the two populations or a common origin of the founders of the two populations. By the analysis of a previously described EST library, we characterized 38 MHC cDNA sequences (17 class I and 21 class II). In conclusion, we characterized for the first time the MHC polymorphism of Barbados and Saint Kitts AGM. We found a restricted polymorphism due to a founding effect, which is responsible for a strong bottleneck. The poorness of MHC polymorphism observed in the Caribbean AGM populations is similar to that observed in the Mauritian cynomolgus macaque population.
Collapse
|
9
|
Iwase H, Ezzelarab MB, Ekser B, Cooper DKC. The role of platelets in coagulation dysfunction in xenotransplantation, and therapeutic options. Xenotransplantation 2014; 21:201-20. [PMID: 24571124 DOI: 10.1111/xen.12085] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 01/08/2014] [Indexed: 12/11/2022]
Abstract
Xenotransplantation could resolve the increasing discrepancy between the availability of deceased human donor organs and the demand for transplantation. Most advances in this field have resulted from the introduction of genetically engineered pigs, e.g., α1,3-galactosyltransferase gene-knockout (GTKO) pigs transgenic for one or more human complement-regulatory proteins (e.g., CD55, CD46, CD59). Failure of these grafts has not been associated with the classical features of acute humoral xenograft rejection, but with the development of thrombotic microangiopathy in the graft and/or consumptive coagulopathy in the recipient. Although the precise mechanisms of coagulation dysregulation remain unclear, molecular incompatibilities between primate coagulation factors and pig natural anticoagulants exacerbate the thrombotic state within the xenograft vasculature. Platelets play a crucial role in thrombosis and contribute to the coagulation disorder in xenotransplantation. They are therefore important targets if this barrier is to be overcome. Further genetic manipulation of the organ-source pigs, such as pigs that express one or more coagulation-regulatory genes (e.g., thrombomodulin, endothelial protein C receptor, tissue factor pathway inhibitor, CD39), is anticipated to inhibit platelet activation and the generation of thrombus. In addition, adjunctive pharmacologic anti-platelet therapy may be required. The genetic manipulations that are currently being tested are reviewed, as are the potential pharmacologic agents that may prove beneficial.
Collapse
Affiliation(s)
- Hayato Iwase
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
10
|
Angiolillo DJ, Ferreiro JL. Antiplatelet and anticoagulant therapy for atherothrombotic disease: the role of current and emerging agents. Am J Cardiovasc Drugs 2013; 13:233-50. [PMID: 23613159 DOI: 10.1007/s40256-013-0022-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Coronary atherothrombotic disease, including chronic stable angina and acute coronary syndromes (ACS), is associated with significant global burden. The acute clinical manifestations of atherothrombotic disease are mediated by occlusive arterial thrombi that impair tissue perfusion and are composed of a core of aggregated platelets, generated by platelet activation, and a superimposed fibrin mesh produced by the coagulation cascade. Long-term antithrombotic therapies, namely oral antiplatelet agents and anticoagulants, have demonstrated variable clinical effects. Aspirin and P2Y12 adenosine diphosphate (ADP) receptor antagonists have been shown to reduce the risk for thrombosis and ischaemic events by blocking the thromboxane (Tx) A2 and platelet P2Y12 activation pathways, respectively, whereas the benefits of oral anticoagulants have not been consistently documented. However, even in the presence of aspirin and a P2Y12 receptor antagonist, the risk for ischaemic events remains substantial because platelet activation continues via pathways independent of TxA2 and ADP, most notably the protease-activated receptor (PAR)-1 platelet activation pathway stimulated by thrombin. Emerging antithrombotic therapies include those targeting the platelet, such as the new P2Y12 antagonists and a novel class of oral PAR-1 antagonists, and those inhibiting the coagulation cascade, such as the new direct factor Xa antagonists, the direct thrombin inhibitors, and a novel class of factor IX inhibitors. The role of emerging antiplatelet agents and anticoagulants in the long-term management of patients with atherothrombotic disease will be determined by the balance of efficacy and safety in large ongoing clinical trials.
Collapse
|
11
|
Olivier C, Diehl P, Bode C, Moser M. Thrombin receptor antagonism in antiplatelet therapy. Cardiol Ther 2013; 2:57-68. [PMID: 25135289 PMCID: PMC4107434 DOI: 10.1007/s40119-013-0013-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Indexed: 12/17/2022] Open
Abstract
Activated platelets play a crucial role in the pathogenesis of atherothrombotic disease and its complications. Even under treatment of antiplatelet drugs, such as acetylsalicylic acid and P2Y12 antagonists, morbidity and mortality rates of thromboembolic complications remain high. Hence, the therapeutic inhibition of protease-activated receptor (PAR)-1, which is activated by thrombin, is a novel promising approach in antiplatelet therapy. Recent data suggest that PAR-1 is mainly involved in pathological thrombus formation, but not in physiological hemostasis. Therefore, PAR-1 inhibition offers the possibility to reduce atherothrombotic events without increasing bleeding risk. So far, two emerging PAR-1 antagonists have been tested in clinical trials: vorapaxar (SCH530349; Merck & Co., Whitehouse Station, NJ, USA) and atopaxar (E5555; Eisai, Tokyo, Japan). Although in TRA-CER vorapaxar showed an unfavorable profile for patients with acute coronary syndrome in addition to standard therapy, it revealed promising results for patients with prior myocardial infarction in TRA 2P-TIMI50. Depending on the status of clinical approval, vorapaxar might be an option for patients with peripheral arterial disease to reduce limb ischemia. The second PAR-I antagonist, atopaxar, tended towards reducing major cardiovascular adverse events in acute coronary syndrome patients in a phase II trial. However, although statistically not significant, bleeding events were numerically increased in atopaxar-treated patients compared with placebo. Furthermore, liver enzymes were elevated and the relative corrected QT interval was prolonged in atopaxar-treated patients. Currently, the development of atopaxar by Eisai is discontinued. The future of this novel class of antithrombotic drugs will depend on the identification of patient groups in which the risk–benefit ratio is favorable.
Collapse
Affiliation(s)
- C Olivier
- Cardiology and Angiology I, Heart Center, Freiburg University, Freiburg, Germany,
| | | | | | | |
Collapse
|
12
|
Capodanno D, Bhatt DL, Goto S, O'Donoghue ML, Moliterno DJ, Tamburino C, Angiolillo DJ. Safety and efficacy of protease-activated receptor-1 antagonists in patients with coronary artery disease: a meta-analysis of randomized clinical trials. J Thromb Haemost 2012; 10:2006-15. [PMID: 22845871 DOI: 10.1111/j.1538-7836.2012.04869.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Thrombin receptor antagonists blocking protease-activated receptor-1 (PAR-1) on platelets represent a new class of oral antiplatelet agents for patients with atherothrombotic disease manifestations. OBJECTIVES We investigated the safety and efficacy of PAR-1 antagonists in patients with coronary artery disease (CAD). PATIENTS/METHODS Randomized, placebo-controlled trials of the PAR-1 antagonists atopaxar or vorapaxar in CAD patients were identified. The primary safety endpoint was the composite of Thrombolysis In Myocardial Infarction (TIMI) clinically significant bleeding. The primary efficacy endpoint was the composite of death, myocardial infarction (MI) or stroke. RESULTS A total of 41 647 patients from eight trials were included. PAR-1 antagonists were associated with higher risks of TIMI clinically significant (odds ratio [OR] 1.48, 95% confidence interval [CI] 1.39-1.57, P < 0.001), major (OR 1.46, 95% CI 1.28-1.67, P < 0.001) and minor (OR 1.67, 95% CI 1.40-2.00, P < 0.001) bleeding than placebo in the fixed-effects model. PAR-1 antagonists reduced the composite of death, MI or stroke as compared with placebo (OR 0.87, 95% CI 0.81-0.92, P < 0.001), driven by a lower risk of MI (OR 0.85, 95% CI 0.78-0.92, P < 0.001). Conversely, PAR-1 antagonists and placebo did not differ in terms of risk of death (OR 0.99, 95% CI 0.90-1.09, P = 0.81) or stroke (OR 0.96, 95% CI 0.84-1.10, P = 0.59). CONCLUSIONS PAR-1 antagonists decrease ischemic events in patients with CAD as compared with placebo, mainly driven by a reduction in MI, at the cost of an increased risk of clinically significant bleeding.
Collapse
|
13
|
Expression sequence tag library derived from peripheral blood mononuclear cells of the chlorocebus sabaeus. BMC Genomics 2012; 13:279. [PMID: 22726727 PMCID: PMC3539953 DOI: 10.1186/1471-2164-13-279] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 06/11/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND African Green Monkeys (AGM) are amongst the most frequently used nonhuman primate models in clinical and biomedical research, nevertheless only few genomic resources exist for this species. Such information would be essential for the development of dedicated new generation technologies in fundamental and pre-clinical research using this model, and would deliver new insights into primate evolution. RESULTS We have exhaustively sequenced an Expression Sequence Tag (EST) library made from a pool of Peripheral Blood Mononuclear Cells from sixteen Chlorocebus sabaeus monkeys. Twelve of them were infected with the Simian Immunodeficiency Virus. The mononuclear cells were or not stimulated in vitro with Concanavalin A, with lipopolysacharrides, or through mixed lymphocyte reaction in order to generate a representative and broad library of expressed sequences in immune cells. We report here 37,787 sequences, which were assembled into 14,410 contigs representing an estimated 12% of the C. sabaeus transcriptome. Using data from primate genome databases, 9,029 assembled sequences from C. sabaeus could be annotated. Sequences have been systematically aligned with ten cDNA references of primate species including Homo sapiens, Pan troglodytes, and Macaca mulatta to identify ortholog transcripts. For 506 transcripts, sequences were quasi-complete. In addition, 6,576 transcript fragments are potentially specific to the C. sabaeus or corresponding to not yet described primate genes. CONCLUSIONS The EST library we provide here will prove useful in gene annotation efforts for future sequencing of the African Green Monkey genomes. Furthermore, this library, which particularly well represents immunological and hematological gene expression, will be an important resource for the comparative analysis of gene expression in clinically relevant nonhuman primate and human research.
Collapse
|
14
|
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]
|
15
|
Targeting proteinase-activated receptors: therapeutic potential and challenges. Nat Rev Drug Discov 2012; 11:69-86. [PMID: 22212680 DOI: 10.1038/nrd3615] [Citation(s) in RCA: 248] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Proteinase-activated receptors (PARs), a family of four seven-transmembrane G protein-coupled receptors, act as targets for signalling by various proteolytic enzymes. PARs are characterized by a unique activation mechanism involving the proteolytic unmasking of a tethered ligand that stimulates the receptor. Given the emerging roles of these receptors in cancer as well as in disorders of the cardiovascular, musculoskeletal, gastrointestinal, respiratory and central nervous system, PARs have become attractive targets for the development of novel therapeutics. In this Review we summarize the mechanisms by which PARs modulate cell function and the roles they can have in physiology and diseases. Furthermore, we provide an overview of possible strategies for developing PAR antagonists.
Collapse
|
16
|
Kogushi M, Matsuoka T, Kawata T, Kuramochi H, Kawaguchi S, Murakami K, Hiyoshi H, Suzuki S, Kawahara T, Kajiwara A, Hishinuma I. The novel and orally active thrombin receptor antagonist E5555 (Atopaxar) inhibits arterial thrombosis without affecting bleeding time in guinea pigs. Eur J Pharmacol 2011; 657:131-7. [PMID: 21300059 DOI: 10.1016/j.ejphar.2011.01.058] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 01/07/2011] [Accepted: 01/25/2011] [Indexed: 10/18/2022]
Abstract
Thrombin is a powerful agonist for platelets, the action of which is mediated by the thrombin receptor protease-activated receptor-1 (PAR-1). Recently, we discovered that E5555 (1-(3-tert-butyl-4-methoxy-5-morpholinophenyl)-2-(5,6-diethoxy-7-fluoro-1-imino-1,3-dihydro-2H-isoindol-2-yl) ethanone hydrobromide) is a potent thrombin receptor antagonist. We evaluated the anti-platelet and anti-thrombotic effects of E5555. E5555 inhibited the binding of a high-affinity thrombin receptor-activating peptide ([(3)H]haTRAP) to PAR-1 with a half maximal inhibitory concentration (IC(50)) value of 0.019μM. E5555 showed potent inhibitory effects on human platelet aggregation induced by thrombin and TRAP with IC(50) values of 0.064 and 0.031μM, respectively, but had no effect on platelet aggregation induced by either ADP or collagen. Similarly, E5555 showed potent and selective inhibitory effects on guinea pig platelet aggregation induced by thrombin and TRAP with IC(50) values of 0.13 and 0.097μM, respectively. The antithrombotic activity of E5555 in vivo was evaluated in a photochemically-induced thrombosis (PIT) model using guinea pigs. Oral administration of E5555 at 30 and 100mg/kg prolonged the time to occlusion by 1.8-fold and 2.4-fold, respectively, compared with controls. Furthermore, E5555 did not prolong bleeding time in guinea pigs at the highest tested dosage of 1000mg/kg. The drug interactions between E5555 and tissue plasminogen activator (tPA) were evaluated. Intravenous administration of 1mg/kg tPA significantly prolonged bleeding time, and its effects were not altered by the oral co-administration of 300mg/kg E5555. These results suggest that E5555 could be a therapeutic option for atherothrombotic disease.
Collapse
Affiliation(s)
- Motoji Kogushi
- Eisai Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki 300–2635, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
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
|
18
|
Chackalamannil S, Xia Y. Thrombin receptor (PAR-1) antagonists as novel antithrombotic agents. Expert Opin Ther Pat 2010; 16:493-505. [PMID: 20144050 DOI: 10.1517/13543776.16.4.493] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
In addition to its central role in haemostasis and wound healing, thrombin activates platelets and smooth muscle cells by proteolytic activation of cell surface protease-activated receptor-1 (PAR-1), which is also known as the thrombin receptor. Thrombin is the most potent activator of human platelets and, as such, a thrombin receptor antagonist is likely to exert potent antithrombotic effect in platelet-rich arterial thrombosis. As thrombin receptor antagonism does not inhibit the ability of thrombin to generate fibrin, such an agent is likely to have less bleeding liability than conventional anticoagulants. The proof-of-concept of the antithrombotic effect of PAR-1 antagonists has been established in several non-human primate models. The current success of PAR-1 research is underscored by the advancement of two candidates into clinical trails for acute coronary syndrome by Schering-Plough and Eisai Company.
Collapse
Affiliation(s)
- Samuel Chackalamannil
- Schering-Plough Research Institute, 2015 Galloping Hill Rd, Kenilworth, NJ 07033, USA.
| | | |
Collapse
|
19
|
Abstract
Clinical manifestations of atherothrombotic disease, such as acute coronary syndromes, cerebrovascular events, and peripheral arterial disease, are major causes of mortality and morbidity worldwide. Platelet activation and aggregation are ultimately responsible for the progression and clinical presentations of atherothrombotic disease. The current standard of care, dual oral antiplatelet therapy with aspirin and the P2Y(12) adenosine diphosphate (ADP) receptor inhibitor clopidogrel, has been shown to improve outcomes in patients with atherothrombotic disease. However, aspirin and P2Y(12) inhibitors target the thromboxane A(2) and the ADP P2Y(12) platelet activation pathways and minimally affect other pathways, while agonists such as thrombin, considered to be the most potent platelet activator, continue to stimulate platelet activation and thrombosis. This may help explain why patients continue to experience recurrent ischaemic events despite receiving such therapy. Furthermore, aspirin and P2Y(12) receptor antagonists are associated with bleeding risk, as the pathways they inhibit are critical for haemostasis. The challenge remains to develop therapies that more effectively inhibit platelet activation without increasing bleeding complications. The inhibition of the protease-activated receptor-1 (PAR-1) for thrombin has been shown to inhibit thrombin-mediated platelet activation without increasing bleeding in pre-clinical models and small-scale clinical trials. PAR-1 inhibition in fact does not interfere with thrombin-dependent fibrin generation and coagulation, which are essential for haemostasis. Thus PAR-1 antagonism coupled with existing dual oral antiplatelet therapy may potentially offer more comprehensive platelet inhibition without the liability of increased bleeding.
Collapse
Affiliation(s)
- Dominick J Angiolillo
- Division of Cardiology, Department of Medicine, University of Florida College of Medicine -Jacksonville, Shands Jacksonville, 655 West 8th St, Jacksonville, FL 32209, USA.
| | | | | |
Collapse
|
20
|
|
21
|
Ward KW, Coon DJ, Magiera D, Bhadresa S, Nisbett E, Lawrence MS. Exploration of the African green monkey as a preclinical pharmacokinetic model: intravenous pharmacokinetic parameters. Drug Metab Dispos 2008; 36:715-20. [PMID: 18227143 DOI: 10.1124/dmd.107.019315] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
The value of cynomolgus and rhesus monkeys to predict human pharmacokinetic parameters has been well established in recent years. However, practical limitations on cost and accessibility can often be a deterrent to obtain data in these valuable species, and the characterization of the predictive power of other nonhuman primates would be useful. Therefore, the present investigation was designed to evaluate the pharmacokinetics of a test set of marketed compounds in the African green monkey, to compare the pharmacokinetics of these agents between nonhuman primate species, and to validate the ability of the African green monkey to predict human pharmacokinetics. Intravenous pharmacokinetics were evaluated for 11 test compounds in this study and compared with data from rats, dogs, cynomolgus/rhesus monkeys, and humans. The results from this investigation indicate that African green monkeys deliver reasonable prediction of human clearance and mean residence time and volume of distribution, although somewhat less accurately than cynomolgus and rhesus monkeys, particularly for volume of distribution, potentially because of body size or composition or experimental design differences. Furthermore, use of an optimized clearance prediction algorithm from the literature enhanced predictivity over a simple liver blood flow-based extrapolation methodology. The data from this study show that African green monkeys have the potential to be used as a surrogate for cynomolgus or rhesus monkeys in preclinical pharmacokinetic studies, particularly for the study of clearance processes, and should be considered as an alternate nonhuman primate test species.
Collapse
|
22
|
Chintala M, Shimizu K, Ogawa M, Yamaguchi H, Doi M, Jensen P. Basic and Translational Research on Proteinase-Activated Receptors: Antagonism of the Proteinase-Activated Receptor 1 for Thrombin, a Novel Approach to Antiplatelet Therapy for Atherothrombotic Disease. J Pharmacol Sci 2008; 108:433-8. [DOI: 10.1254/jphs.08r06fm] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
23
|
Hamilton JR, Kaplan Z, Jackson SP, Opat S. Antiplatelet therapy: present status and future prospects. Expert Opin Drug Discov 2007; 2:1035-40. [DOI: 10.1517/17460441.2.8.1035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Justin R Hamilton
- Australian Centre for Blood Diseases, Monash University, 6th Level, Burnet Building, AMREP Campus, 89 Commercial Road, Melbourne, Victoria 3004, Australia ;
| | - Zane Kaplan
- Department of Haematology, Alfred Hospital, Melbourne, Victoria 3004, Australia
| | - Shaun P Jackson
- Australian Centre for Blood Diseases, Monash University, 6th Level, Burnet Building, AMREP Campus, 89 Commercial Road, Melbourne, Victoria 3004, Australia ;
| | - Stephen Opat
- Clinical Haematology Unit, Monash Medical Centre, Clayton, Victoria 3168, Australia
| |
Collapse
|
24
|
Landis RC. Protease activated receptors: clinical relevance to hemostasis and inflammation. Hematol Oncol Clin North Am 2007; 21:103-13. [PMID: 17258121 DOI: 10.1016/j.hoc.2006.11.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The protease-activated receptors (PARs) are a unique family of vascular receptors that confer on cells an ability to sense, and respond to, local changes in the proteolytic environment. They are activated by serine proteases of the blood coagulation cascade, notably thrombin, and are linked to thrombotic and inflammatory effector pathways. In surgery with cardiopulmonary bypass (CPB), thrombin is generated in large quantities in the extracorporeal circuit and can exert systemic effects by way of platelet and endothelial PAR1. Aprotinin (Trasylol), a serine protease inhibitor used in cardiac surgery, preserves platelet function, and attenuates the inflammatory response by protecting the PAR 1 receptor on platelets and endothelium.
Collapse
Affiliation(s)
- R Clive Landis
- Edmund Cohen Laboratory for Vascular Research, University of the West Indies, Chronic Disease Research Centre, Jemmotts Lane, Barbados, West Indies.
| |
Collapse
|
25
|
Bahou WF. Thrombin Receptors. Platelets 2007. [DOI: 10.1016/b978-012369367-9/50771-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
|
26
|
Vandendries ER, Hamilton JR, Coughlin SR, Furie B, Furie BC. Par4 is required for platelet thrombus propagation but not fibrin generation in a mouse model of thrombosis. Proc Natl Acad Sci U S A 2006; 104:288-92. [PMID: 17190826 PMCID: PMC1765451 DOI: 10.1073/pnas.0610188104] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Thrombin, a central mediator of hemostasis and thrombosis, converts fibrinogen to fibrin and is a potent platelet activator. Activated platelets provide a surface for assembly of the tenase and prothrombinase complexes required for thrombin generation. The role of thrombin-induced platelet activation in platelet accumulation and its interplay with fibrin deposition during thrombus assembly has not been fully defined. We studied these processes during laser-induced thrombus formation by using real-time digital fluorescence microscopy in mice lacking protease-activated receptor-4 (Par4), which is necessary for thrombin responsiveness in mouse platelets. Juxtamural platelet accumulation immediately after laser injury was not different in wild-type and Par4(-/-) mice. However, subsequent growth of platelet thrombi was markedly diminished in Par4(-/-) mice. At the time of maximal thrombus size in wild type, platelet accumulation was more than 10-fold higher in wild type than in Par4(-/-) mice. P-selectin expression, a marker of platelet activation, was reduced and delayed in Par4(-/-) thrombi. In contrast to platelet activation and accumulation, the rate and amount of fibrin deposition, predominantly intramural and juxtamural in this model, were indistinguishable in Par4(-/-) and wild-type mice. These results suggest that platelet activation by thrombin is necessary for normal propagation of a platelet thrombus at a distance from the injured vessel wall and hence for normal thrombus growth. However, platelet activation by thrombin is unnecessary for initial and limited accumulation of platelets at or near the vessel wall, and this limited accumulation of platelets and/or platelet-independent mechanism(s) of thrombin generation are sufficient for normal fibrin deposition in this model.
Collapse
Affiliation(s)
- Erik R. Vandendries
- *Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215; and
| | - Justin R. Hamilton
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143
| | - Shaun R. Coughlin
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143
- To whom correspondence may be addressed. E-mail:
| | - Bruce Furie
- *Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215; and
| | - Barbara C. Furie
- *Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215; and
- To whom correspondence may be addressed at:
Beth Israel Deaconess Medical Center, 840 Memorial Drive, Cambridge, MA 02139. E-mail:
| |
Collapse
|
27
|
Wu CC, Teng CM. Comparison of the effects of PAR1 antagonists, PAR4 antagonists, and their combinations on thrombin-induced human platelet activation. Eur J Pharmacol 2006; 546:142-7. [PMID: 16890935 DOI: 10.1016/j.ejphar.2006.07.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2005] [Revised: 05/26/2006] [Accepted: 07/07/2006] [Indexed: 10/24/2022]
Abstract
Thrombin activates human platelets through proteolytic activation of two protease-activated receptors (PARs), PAR1 and PAR4. In the present study, we show that, RWJ-56110, a potent synthetic PAR1 antagonist, inhibited platelet aggregation caused by a low concentration (0.05 U/ml) of thrombin, but lost its effectiveness when higher concentrations of thrombin were used as stimulators. YD-3, a non-peptide PAR4 antagonist, alone had little or no effect on thrombin-induced platelet aggregation, significantly enhanced the anti-aggregatory activity of PAR1 antagonist. In addition, we demonstrate for the first time that P-selectin expression in thrombin-stimulated platelets can be synergistically prevented by combined treatment of PAR1 antagonist and PAR4 antagonist. These results indicate that thrombin-induced platelet activation cannot be effectively inhibited by just blocking either single thrombin receptor pathway, and suggest a rationale for potential combination therapy in arterial thrombosis.
Collapse
Affiliation(s)
- Chin-Chung Wu
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | | |
Collapse
|
28
|
Abstract
The coagulation cascade and protease-activated receptors (PARs) together provide an elegant mechanism that links mechanical information in the form of tissue injury to cellular responses. These receptors appear to largely account for the cellular effects of thrombin and can mediate signaling to other trypsin-like proteases. An important role for PARs in hemostasis and thrombosis is established in animal models, and studies in knockout mice and nonhuman primates raise the question of whether PAR inhibition might offer an appealing new approach to the prevention and treatment of thrombosis. PARs may also trigger inflammatory responses to tissue injury. For example, PAR activation on endothelial cells and perhaps sensory afferents can trigger local accumulation of leukocytes and platelets and transudation of plasma. However, panoply of signaling systems and cell types orchestrates inflammatory responses, and efforts to define the relative importance and roles of PARs in various inflammatory processes are just beginning. Lastly, roles for PARs in blood vessel formation and other processes during embryonic development are emerging, and whether these reflect new roles for the coagulation cascade and/or PAR signaling to other proteases remains to be explored.
Collapse
Affiliation(s)
- S R Coughlin
- Cardiovascular Research Institute, Department of Medicine, University of California, San Francisco, CA 94143-0130, USA.
| |
Collapse
|
29
|
Steinhoff M, Buddenkotte J, Shpacovitch V, Rattenholl A, Moormann C, Vergnolle N, Luger TA, Hollenberg MD. Proteinase-activated receptors: transducers of proteinase-mediated signaling in inflammation and immune response. Endocr Rev 2005; 26:1-43. [PMID: 15689571 DOI: 10.1210/er.2003-0025] [Citation(s) in RCA: 369] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Serine proteinases such as thrombin, mast cell tryptase, trypsin, or cathepsin G, for example, are highly active mediators with diverse biological activities. So far, proteinases have been considered to act primarily as degradative enzymes in the extracellular space. However, their biological actions in tissues and cells suggest important roles as a part of the body's hormonal communication system during inflammation and immune response. These effects can be attributed to the activation of a new subfamily of G protein-coupled receptors, termed proteinase-activated receptors (PARs). Four members of the PAR family have been cloned so far. Thus, certain proteinases act as signaling molecules that specifically regulate cells by activating PARs. After stimulation, PARs couple to various G proteins and activate signal transduction pathways resulting in the rapid transcription of genes that are involved in inflammation. For example, PARs are widely expressed by cells involved in immune responses and inflammation, regulate endothelial-leukocyte interactions, and modulate the secretion of inflammatory mediators or neuropeptides. Together, the PAR family necessitates a paradigm shift in thinking about hormone action, to include proteinases as key modulators of biological function. Novel compounds that can modulate PAR function may be potent candidates for the treatment of inflammatory or immune diseases.
Collapse
Affiliation(s)
- Martin Steinhoff
- Department of Dermatology and Boltzmann Institute for Immunobiology of the Skin, University of Münster, von-Esmarch-Strasse 58, 48149 Münster, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Nieman MT, Warnock M, Hasan AAK, Mahdi F, Lucchesi BR, Brown NJ, Murphey LJ, Schmaier AH. The preparation and characterization of novel peptide antagonists to thrombin and factor VIIa and activation of protease-activated receptor 1. J Pharmacol Exp Ther 2004; 311:492-501. [PMID: 15210836 DOI: 10.1124/jpet.104.069229] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Thrombin and protease-activated receptor 1 (PAR1) activation antagonists were prepared based upon the peptide RPPGF, the angiotensin-converting enzyme breakdown product of bradykinin. A library of 72 peptides consisting of d and/or synthetic amino acids was designed with various substitutions in positions 1 to 5 in Arg-Pro-Pro-Gly-Phe (RPPGF). Two compounds, rOicPGF (TH146) and betaAK2K-4(rOicPGF) (MAP4-TH146), were characterized further. TH146 or MAP4-TH146 completely inhibits threshold gamma-thrombin-induced platelet aggregation at a concentration of 142 +/- 0.05 or 19 +/- 0.06 microM, respectively. TH146 completely inhibits threshold alpha-thrombin-induced washed platelet aggregation at 444 +/- 0.04 microM. TH146 or MAP4-TH146 blocks 2 nM alpha-thrombin-induced fibroblast calcium mobilization with an IC(50) value of 110 or 18 microM, respectively. Furthermore, significant prolongation of the activated partial thromboplastin time, prothrombin time, or thrombin clotting time occurs at 31, 62, or 7.8 microM TH146 and 0.4, 6.25, or 1.56 microM MAP4-TH146, respectively. TH146 and MAP4-TH146 inhibit both alpha-thrombin with a K(i) value of 97 and 49 microM, respectively, and factor VIIa with a K(i) value of 44 and 5 microM, respectively. Both TH146 and MAP4-TH146 specifically bind to the exodomain of recombinant PAR1. MAP4-TH146 (200 microM) completely blocks thrombocytin, a PAR1-activating snake venom protease, without inhibiting the enzyme's active site. TH146 inhibits gamma-thrombin-induced aggregation of mouse platelets, prolongs mouse bleeding times, and delays the time to mouse carotid artery thrombosis. TH146 and MAP4-TH146 inhibit human and mouse platelet aggregation and mouse thrombosis. Analogs of RPPGF are model compounds to develop PAR1 activation antagonists as well as direct inhibitors to thrombin and factor VIIa.
Collapse
|
31
|
Hamilton JR, Cornelissen I, Coughlin SR. Impaired hemostasis and protection against thrombosis in protease-activated receptor 4-deficient mice is due to lack of thrombin signaling in platelets. J Thromb Haemost 2004; 2:1429-35. [PMID: 15304051 DOI: 10.1111/j.1538-7836.2004.00783.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Platelets from protease-activated receptor 4 (PAR4)-deficient mice are unresponsive to thrombin, and Par4-/- mice have prolonged bleeding times and are protected against thrombosis. However, in addition to its role in platelets, PAR4 contributes to thrombin signaling in cells in the blood vessel wall that might participate in hemostasis and thrombosis, such as endothelial cells. To determine whether the hemostatic and thrombotic phenotypes of Par4-/- mice were due to loss of PAR4 function in hematopoietic vs. other cell types, tail bleed times and thromboplastin-induced pulmonary embolism were examined in lethally irradiated mice reconstituted with Par4+/+ or Par4-/- bone marrow. In Par4+/+ and Par4-/- mice reconstituted with Par4+/+ marrow, the median tail bleed times were 2.0 and 1.7 min, respectively, vs. > 10 min for both Par4+/+ and Par4-/- mice reconstituted with Par4-/- marrow. In the pulmonary embolism model, Par4+/+ and Par4-/- mice reconstituted with Par4+/+ marrow survived a median of 3.7 and 2.8 min, respectively, after administration of thromboplastin, vs. > 20 min for both Par4+/+ and Par4-/- mice reconstituted with Par4-/- marrow. Further, the phenotype of mice reconstituted with Par4-/- marrow was almost as dramatic as that seen in Nf-e2-/- mice, which lack platelets. These data strongly suggest that increased tail bleed times and protection against thrombosis in Par4-/- mice are accounted for by lack of PAR4 function in platelets, emphasize the importance of thrombin signaling in platelets among the multiple pathways and cell types that govern hemostasis and thrombosis.
Collapse
Affiliation(s)
- J R Hamilton
- Cardiovascular Research Institute, University of California, San Francisco, USA
| | | | | |
Collapse
|
32
|
Affiliation(s)
- Wadie F Bahou
- Division of Hematology, State University of New York, Stony Brook, New York 11794-8151, USA
| |
Collapse
|
33
|
Seeley S, Covic L, Jacques SL, Sudmeier J, Baleja JD, Kuliopulos A. Structural Basis for Thrombin Activation of a Protease-Activated Receptor. ACTA ACUST UNITED AC 2003; 10:1033-41. [PMID: 14652070 DOI: 10.1016/j.chembiol.2003.10.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Protease-activated G protein-coupled receptors (PAR1-4) are tethered-ligand receptors that are activated by proteolytic cleavage of the extracellular domain (exodomain) of the receptor. PAR1, the prototypic member of the PAR family, is the high-affinity thrombin receptor of platelets and vascular endothelium and plays a critical role in blood coagulation, thrombosis, and inflammation. Here, we describe the solution structure of the thrombin-cleaved exodomain of PAR1. The side chains of a hydrophobic hirudin-like (Hir) sequence and adjacent anionic motif project into solution. Docking of the exodomain Hir sequence to exosite I of thrombin reveals that the tethered ligand in the cleaved exodomain bends away from thrombin, leaving its active site available to another large macromolecular substrate. The N-terminal ligand is longer than anticipated and forms an intramolecular complex with a region located in the C terminus of the exodomain. Mutational analysis confirmed that this C-terminal region is a ligand binding site for both intra- and intermolecular ligands. A lipidated-ligand binding site peptide was found to be an effective inhibitor of thrombin-induced platelet aggregation.
Collapse
Affiliation(s)
- Stacy Seeley
- Division of Hematology/Oncology and Department of Medicine, New England Medical Center, Boston, MA 02111, USA
| | | | | | | | | | | |
Collapse
|
34
|
Coughlin SR. Protease-activated receptors in the cardiovascular system. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2003; 67:197-208. [PMID: 12858541 DOI: 10.1101/sqb.2002.67.197] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- S R Coughlin
- Cardiovascular Research Institute, Department of Medicine and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California 94143, USA
| |
Collapse
|
35
|
Affiliation(s)
- Greg Stratmann
- *Department of Anesthesia and Perioperative Care and †Pediatrics, University of California at San Francisco
| | | |
Collapse
|
36
|
Affiliation(s)
- Shaun R Coughlin
- Cardiovascular Research Institute, University of California at San Francisco, San Francisco, California 94143, USA.
| | | |
Collapse
|
37
|
Dupont A, Fontana P, Bachelot-Loza C, Reny JL, Biéche I, Desvard F, Aiach M, Gaussem P. An intronic polymorphism in the PAR-1 gene is associated with platelet receptor density and the response to SFLLRN. Blood 2003; 101:1833-40. [PMID: 12406873 DOI: 10.1182/blood-2002-07-2149] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Protease-activated receptor 1 (PAR-1), the main thrombin receptor on vascular cells, plays a key role in platelet activation. We examined the range of PAR-1 expression on platelets, obtained twice, 1 week apart, from 100 healthy subjects and found a 2-fold interindividual variation in receptor numbers (95% CI = 858-1700). Because PAR-1 density was stable with time (r(2) = 76%, P <.001), we sought a genetic explanation for the observed variability. To validate this approach, we also analyzed the alpha(2)beta(1) genotype according to receptor density and platelet mRNA expression data. We found that the number of PAR-1 receptors on the platelet surface is associated with the intervening sequence IVSn-14 A/T intronic variation. The number of receptors was also found to govern the platelet response to the SFLLRN agonist, in terms of aggregation and P-selectin expression. The T allele (allelic frequency, 0.14) can be considered as an allele with decreased expression, because it was associated with lower PAR-1 expression on the platelet surface and with a lower response to SFLLRN. The IVSn-14 A/T intronic variation may therefore be clinically relevant.
Collapse
Affiliation(s)
- Annabelle Dupont
- Service d'Hématologie Biologique and INSERM Unité 428, Hôpital Européen Georges Pompidou, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris V, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Derian CK, Damiano BP, Addo MF, Darrow AL, D'Andrea MR, Nedelman M, Zhang HC, Maryanoff BE, Andrade-Gordon P. Blockade of the thrombin receptor protease-activated receptor-1 with a small-molecule antagonist prevents thrombus formation and vascular occlusion in nonhuman primates. J Pharmacol Exp Ther 2003; 304:855-61. [PMID: 12538843 DOI: 10.1124/jpet.102.042663] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Although it is well recognized that human platelet responses to alpha-thrombin are mediated by the protease-activated receptors PAR-1 and PAR-4, their role and relative importance in platelet-dependent human disease has not yet been elucidated. Because the expression profile of PARs in platelets from nonprimates differs from humans, we used cynomolgus monkeys to evaluate the role of PAR-1 in thrombosis. Based on reverse transcription-polymerase chain reaction, PAR expression in platelets from cynomolgus monkeys consisted primarily of PAR-1 and PAR-4, thereby mirroring the profile of human platelets. We probed the role of PAR-1 in a primate model of vascular injury-induced thrombosis with the selective PAR-1 antagonist (alpha S)-N-[(1S)-3-amino-1-[[(phenylmethyl)amino]carbonyl]propyl]-alpha-[[[[[1-(2,6-dichlorophenyl)methyl]-3-(1-pyrrolidinylmethyl)-1H-indazol-6-yl]amino]carbonyl]amino]-3,4-difluorobenzenepropanamide (RWJ-58259). After pretreatment with RWJ-58259 or vehicle, both carotid arteries of anesthetized monkeys were electrolytically injured and blood flow was monitored for 60 min. Time to occlusion was significantly extended after RWJ-58259 administration (27 +/- 3 to 53 +/- 8 min; p < 0.048). Vessels from three of the five treated animals remained patent. Ex vivo platelet aggregation measurements indicated complete PAR-1 inhibition, as well as an operational PAR-4 response. Immunohistochemical staining of mural thrombi with antibodies to the platelet marker CD61 and fibrinogen indicated that RWJ-58259 significantly reduced thrombus platelet deposition. Drug treatment had no effect on key hematological or coagulation parameters. Our results provide direct evidence that PAR-1 is the primary receptor that mediates alpha-thrombin's prothrombotic actions in primates and suggest that PAR-1 antagonists may have potential for the treatment of thrombotic disorders in humans.
Collapse
Affiliation(s)
- Claudia K Derian
- Johnson & Johnson Pharmaceutical Research and Development, Spring House, Pennsylvania 19477, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Derian CK, Maryanoff BE, Zhang HC, Andrade-Gordon P. Therapeutic potential of protease-activated receptor-1 antagonists. Expert Opin Investig Drugs 2003; 12:209-21. [PMID: 12556215 DOI: 10.1517/13543784.12.2.209] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The serine protease thrombin (EC 3.4.21.5) is central to the maintenance of haemostatic balance through its coagulant, anticoagulant and platelet activating properties. In addition, this enzyme affects numerous cellular responses in a wide variety of cells, such as cell proliferation, cytokine and growth factor release, lipid metabolism and tissue remodelling. A family of G-protein-coupled protease-activated receptors (PARs) mediates these cellular actions of thrombin. While thrombin can activate three of the four PAR family members, PAR-1 represents the primary thrombin-responsive receptor in human cells. The expression of PAR-1 in platelets, the vasculature and myocardium, in cells within atherosclerotic plaque and tissues after vascular injury, indicates that this receptor plays an important role during the response to tissue injury and associated inflammatory processes. With the development of PAR-deficient mice and small-molecule antagonists, it is now clear that intervening in processes mediated by PAR-1 presents a new approach to treating a variety of disorders dependent on thrombin generation, including thrombosis and restenosis. The full potential of PAR-1 antagonists has yet to be realised, but the promise of novel therapeutics that modulate receptor function rather than thrombin's proteolytic activity, provides an alternative and, perhaps, more desirable means to dampen the pathological effects of thrombin.
Collapse
Affiliation(s)
- Claudia K Derian
- Drug Discovery, Johnson & Johnson Pharmaceutical Research & Development, Spring House, Pennsylvania 19477-0776, USA
| | | | | | | |
Collapse
|
40
|
Affiliation(s)
- Shaun R Coughlin
- Cardiovascular Research Institute, University of California at San Francisco, San Francisco, California 94143, USA.
| | | |
Collapse
|
41
|
Abstract
The recent observation that knock-out of protease-activated receptor-4 (PAR4) ablates thrombin signaling in mouse platelets and protects against ferric chloride-induced thrombosis of mouse mesenteric arterioles suggests that thrombin's actions on platelets can play an important role in thrombosis. Complete ablation of thrombin signaling would be difficult to achieve in human beings because human platelets have 2 thrombin receptors that are each capable of mediating transmembrane signaling. However, it is possible that complete ablation of thrombin signaling in platelets is not necessary for an antithrombotic effect. In mouse platelets, PAR3 functions as a cofactor that binds thrombin and promotes productive cleavage of PAR4, and thrombin responses are decreased but not absent in Par3(-/-) platelets. We now report that Par3(-/-) mice were protected against ferric chloride-induced thrombosis of mesenteric arterioles and against thromboplastin-induced pulmonary embolism. Surprisingly, Par3(-/-) and Par4(-/-) mice showed similar degrees of protection in these models and similar prolongation of tail bleeding times. Thus, even a partial decrease in mouse platelet responsiveness to thrombin protected against thrombosis and impaired hemostasis in some settings. These results demonstrate the importance of PAR3's unusual cofactor function and underscore the relative importance of thrombin's actions on platelets in vivo. They also suggest that PAR inhibition might be explored for the prevention or treatment of thrombosis in human beings.
Collapse
Affiliation(s)
- Ethan J Weiss
- Cardiovascular Research Institute, Department of Medicine, University of California-San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143-0130, USA
| | | | | | | |
Collapse
|
42
|
|
43
|
Nantermet PG, Barrow JC, Lundell GF, Pellicore JM, Rittle KE, Young M, Freidinger RM, Connolly TM, Condra C, Karczewski J, Bednar RA, Gaul SL, Gould RJ, Prendergast K, Selnick HG. Discovery of a nonpeptidic small molecule antagonist of the human platelet thrombin receptor (PAR-1). Bioorg Med Chem Lett 2002; 12:319-23. [PMID: 11814787 DOI: 10.1016/s0960-894x(01)00745-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The synthesis and biological evaluation of a series of nonpeptidic small molecule antagonists of the human platelet thrombin receptor (PAR-1) are described. Optimization of the 5-amino-3-arylisoxazole lead resulted in an approximate 100-fold increase in potency. The most potent of these compounds (54) inhibits platelet activation with IC(50)s of 90 nM against the thrombin receptor agonist peptide (TRAP) and 510 nM against thrombin as the agonist. Further, antagonist 54 fully blocks platelet aggregation stimulated by 1 nM thrombin for 10 min.
Collapse
Affiliation(s)
- Philippe G Nantermet
- Department of Medicinal Chemistry, Merck Research Laboratories, West Point, PA 19486, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Thromboembolic diseases remain the main cause of death in Western societies despite current antithrombotic treatments. Recent advances in the molecular bases of haemostasis have highlighted new targets for novel antiplatelet or anticoagulant agents. Considering antiplatelet agents, selective antagonists of specific receptors (von Willebrand factor, collagen or thrombin receptors) are effective in thrombosis models and direct ADP receptor antagonists and nitric-oxide-releasing aspirin are in Phase I-II clinical trials. Concerning anticoagulants, inhibitors of tissue-factor-induced clotting activation, selective inhibitors of thrombin and factor Xa, and components of the anticoagulant protein C system (recombinant activated human protein C or human soluble thrombomodulin) have been studied. Some of these agents have had promising results in Phase III studies. Several achievements are anticipated from the development of new antithrombotics, including a further reduction of cardiovascular mortality and unwanted bleeding, and easier patient management.
Collapse
Affiliation(s)
- Paolo Gresele
- Division of Internal and Cardiovascular Medicine, Dept of Internal Medicine, University of Perugia, Via E. Dal Pozzo, I-06126, Perugia, Italy.
| | | |
Collapse
|
45
|
Barrow JC, Nantermet PG, Selnick HG, Glass KL, Ngo PL, Young MB, Pellicore JM, Breslin MJ, Hutchinson JH, Freidinger RM, Condra C, Karczewski J, Bednar RA, Gaul SL, Stern A, Gould R, Connolly TM. Discovery and initial structure-activity relationships of trisubstituted ureas as thrombin receptor (PAR-1) antagonists. Bioorg Med Chem Lett 2001; 11:2691-6. [PMID: 11591503 DOI: 10.1016/s0960-894x(01)00538-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Thrombin is the most potent agonist of platelet activation, and its effects are predominantly mediated by platelet thrombin receptors. Therefore, antagonists of the thrombin receptor have potential utility for the treatment of thrombotic disorders. Screening of combinatorial libraries revealed 2 to be a potent antagonist of the thrombin receptor. Modifications of this structure produced 11k, which inhibits thrombin receptor stimulated secretion and aggregation of platelets.
Collapse
Affiliation(s)
- J C Barrow
- Department of Medicinal Chemistry, Merck Research Laboratories, West Point, PA 19486, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Sambrano GR, Weiss EJ, Zheng YW, Huang W, Coughlin SR. Role of thrombin signalling in platelets in haemostasis and thrombosis. Nature 2001; 413:74-8. [PMID: 11544528 DOI: 10.1038/35092573] [Citation(s) in RCA: 379] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Platelets are critical in haemostasis and in arterial thrombosis, which causes heart attacks and other events triggered by abnormal clotting. The coagulation protease thrombin is a potent activator of platelets ex vivo. However, because thrombin also mediates fibrin deposition and because multiple agonists can trigger platelet activation, the relative importance of platelet activation by thrombin in haemostasis and thrombosis is unknown. Thrombin triggers cellular responses at least in part through protease-activated receptors (PARs). Mouse platelets express PAR3 and PAR4 (ref. 9). Here we show that platelets from PAR4-deficient mice failed to change shape, mobilize calcium, secrete ATP or aggregate in response to thrombin. This result demonstrates that PAR signalling is necessary for mouse platelet activation by thrombin and supports the model that mouse PAR3 (mPAR3) does not by itself mediate transmembrane signalling but instead acts as a cofactor for thrombin cleavage and activation of mPAR4 (ref. 10). Importantly, PAR4-deficient mice had markedly prolonged bleeding times and were protected in a model of arteriolar thrombosis. Thus platelet activation by thrombin is necessary for normal haemostasis and may be an important target in the treatment of thrombosis.
Collapse
Affiliation(s)
- G R Sambrano
- Cardiovascular Research Institute, University of California, 513 Parnassus Avenue, San Francisco, California 94143-0130, USA
| | | | | | | | | |
Collapse
|
47
|
Ahn HS, Foster C, Boykow G, Stamford A, Manna M, Graziano M. Inhibition of cellular action of thrombin by N3-cyclopropyl-7-[[4-(1-methylethyl)phenyl]methyl]-7H-pyrrolo[3, 2-f]quinazoline-1,3-diamine (SCH 79797), a nonpeptide thrombin receptor antagonist. Biochem Pharmacol 2000; 60:1425-34. [PMID: 11020444 DOI: 10.1016/s0006-2952(00)00460-3] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A growing body of evidence suggests an important contribution of the cellular actions of thrombin to thrombosis and restenosis following angioplasty. Recently we reported on SCH 79797 (N3-cyclopropyl-7-¿[4-(1-methylethyl)phenyl]methyl¿-7H-pyrrolo[3, 2-f]quinazoline-1,3-diamine) and its analogs as new potent, nonpeptide thrombin receptor antagonists. This study further characterizes the biochemical and pharmacological actions of pyrroloquinazoline inhibitors of protease activated receptor-1 (PAR-1) in human platelets and coronary artery smooth muscle cells (hCASMC). SCH 79797 and its N-methyl analog (SCH 203099) inhibited binding of a high-affinity thrombin receptor-activating peptide ([(3)H]haTRAP, Ala-Phe(p-F)-Arg-ChA-HArg-[(3)H]Tyr-NH(2)) to PAR-1 with IC(50) values of 70 and 45 nM, respectively. SCH 79797 inhibited [(3)H]haTRAP binding in a competitive manner. SCH 79797 and SCH 203099 inhibited alpha-thrombin- and haTRAP-induced aggregation of human platelets, but did not inhibit human platelet aggregation induced by the tethered ligand agonist for protease-activated receptor-4 (PAR-4), gamma-thrombin, ADP, or collagen. SCH 203099 inhibited surface expression of P-selectin induced by haTRAP and thrombin, and it did not increase P-selectin expression or prevent thrombin cleavage of the receptor. Thrombin and TFLLRNPNDK-NH(2) (TK), a PAR-1-selective agonist, produced transient increases in cytosolic free Ca(2+) concentration ([Ca(2+)](i)) in hCASMC. This increase in [Ca(2+)](i) was inhibited effectively by SCH 79797. However, the Ca(2+) transients induced by SLIGKV-NH(2,) a PAR-2-selective agonist, were not inhibited by SCH 79797. Thrombin- and TK-stimulated [(3)H]thymidine incorporation also was inhibited completely by SCH 79797. The results of this study demonstrate that SCH 79797 and SCH 203099 are potent, selective antagonists of PAR-1 in human platelets and hCASMC. These data also suggest that the thrombin stimulation of Ca(2+) transients and mitogenesis in hCASMC is mediated primarily through activation of PAR-1.
Collapse
Affiliation(s)
- H S Ahn
- Schering-Plough Research Institute, Kenilworth, NJ, 07033, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Pakala R, Liang CT, Benedict CR. Inhibition of arterial thrombosis by a peptide ligand of the thrombin receptor. Thromb Res 2000; 100:89-96. [PMID: 11053621 DOI: 10.1016/s0049-3848(00)00307-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Thrombin plays an important role in promoting arterial thrombosis by platelet activation and by catalyzing fibrin formation. Use of thrombin inhibitors that block both the platelet-activating and fibrin formation properties of thrombin are associated with hemostasis. This problem might be overcome by developing agents that block only the platelet-activating property of thrombin. Because the platelet-activating property of thrombin is mediated by the thrombin receptor, antagonists of the thrombin receptor might be efficacious and potentially safer with regard to bleeding complications. We investigated whether a peptide ligand (AFLARAA) of the thrombin receptor that blocked alpha-thrombin and thrombin receptor activating peptide-induced platelet aggregation could inhibit thrombosis. A partially occlusive thrombus was generated by application of electric current in rabbit carotid artery. In control animals, the artery was completely occluded within 42+/-12 min after the current was discontinued. When the thrombin receptor activating peptide antagonist was given (100 micromol/kg as an IV bolus followed by 900 micromol/kg infusion for a period of 180 min) starting at the time the current was stopped, blood flow remained patent throughout the infusion period and for an additional 60 min after the infusion was stopped. The antithrombotic effect of the antagonist peptide was not associated with increased bleeding tendency, as judged by the amount of blood adsorbed by a gauze pad placed in a surgical incision extending to the muscle tissue and by a standard template bleeding time. These results indicate that thrombin receptor antagonist peptides can be used as antithrombotic agents.
Collapse
Affiliation(s)
- R Pakala
- Department of Internal Medicine, Division of Cardiology, University of Texas Health Science Center-Medical School, Houston, TX 77030, USA
| | | | | |
Collapse
|
49
|
Abstract
How does a protease act like a hormone to regulate cellular functions? The coagulation protease thrombin (EC 3.4.21.5) activates platelets and regulates the behavior of other cells by means of G protein-coupled protease-activated receptors (PARs). PAR1 is activated when thrombin binds to and cleaves its amino-terminal exodomain to unmask a new receptor amino terminus. This new amino terminus then serves as a tethered peptide ligand, binding intramolecularly to the body of the receptor to effect transmembrane signaling. The irreversibility of PAR1's proteolytic activation mechanism stands in contrast to the reversible ligand binding that activates classical G protein-coupled receptors and compels special mechanisms for desensitization and resensitization. In endothelial cells and fibroblasts, activated PAR1 rapidly internalizes and then sorts to lysosomes rather than recycling to the plasma membrane as do classical G protein-coupled receptors. This trafficking behavior is critical for termination of thrombin signaling. An intracellular pool of thrombin receptors refreshes the cell surface with naïve receptors, thereby maintaining thrombin responsiveness. Thus cells have evolved a trafficking solution to the signaling problem presented by PARs. Four PARs have now been identified. PAR1, PAR3, and PAR4 can all be activated by thrombin. PAR2 is activated by trypsin and by trypsin-like proteases but not by thrombin. Recent studies with knockout mice, receptor-activating peptides, and blocking antibodies are beginning to define the role of these receptors in vivo.
Collapse
Affiliation(s)
- S R Coughlin
- Cardiovascular Research Institute, Department of Medicine, University of California, San Francisco, CA 94143-0130, USA.
| |
Collapse
|
50
|
Kawano K, Ikeda Y, Kondo K, Umemura K. Superiority of platelet integrin GPIIb-IIIa receptor antagonist over aspirin in preventing cyclic flow reductions in the guinea pig middle cerebral artery. Eur J Pharmacol 1999; 374:377-85. [PMID: 10422782 DOI: 10.1016/s0014-2999(99)00305-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We established a photothrombotic occlusion model in the guinea pig middle cerebral artery. In this model, the middle cerebral artery was recanalized within 10 to 20 min after thrombotic occlusion, with subsequent cyclic flow reductions. Cyclic flow reductions in the middle cerebral artery are expected to manage cerebral infarction by modulating arterial patency. Therefore, we evaluated the effect of several antiplatelet agents on the frequency of cyclic flow reductions and subsequent cerebral infarction using this model. A platelet integrin GPIIb-IIIa receptor antagonist, ME3277 (sodium hydrogen [4-[(4,5,6,7-tetrahydrothieno[3,2c]pyridin-2-yl) carbonylamino] acetyl-o-phenylene] dioxydiacetate, 0.3, 1 and 3 mg/kg i.v.) dose-dependently inhibited the ex vivo platelet aggregation induced by ADP (5 microM), collagen (0.8 and 20 microg/ml) and arachidonic acid (100 microM). While the same doses of ME3277 reduced the frequency of the cyclic flow reductions and increased the total patency time of the middle cerebral artery, time to thrombotic occlusion was prolonged only at the highest dose, 3 mg/kg. ME3277 (0.3-3 mg/kg) significantly reduced the infarct volume and improved the neurological deficit at 24 h. In contrast, aspirin (30 mg/kg) did not affect these variables in spite of complete inhibition of platelet aggregation induced by arachidonic acid and collagen (0.8 microg/ml). A thromboxane A2 synthetase inhibitor, sodium ozagrel, significantly increased the total patency time and reduced the infarct volume at 30 mg/kg. Inhibition of prostaglandin I2 generation could explain the effectiveness of sodium ozagrel but not aspirin in this model. These results suggest that platelet integrin GPIIb-IIIa receptor antagonists are more beneficial than aspirin for the treatment of cerebral thrombosis.
Collapse
Affiliation(s)
- K Kawano
- Department of Pharmacology, Hamamatsu University School of Medicine, Japan.
| | | | | | | |
Collapse
|