1
|
Navarro S, Talucci I, Göb V, Hartmann S, Beck S, Orth V, Stoll G, Maric HM, Stegner D, Nieswandt B. The humanized platelet glycoprotein VI Fab inhibitor EMA601 protects from arterial thrombosis and ischaemic stroke in mice. Eur Heart J 2024:ehae482. [PMID: 39150906 DOI: 10.1093/eurheartj/ehae482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/12/2024] [Accepted: 07/12/2024] [Indexed: 08/18/2024] Open
Abstract
BACKGROUND AND AIMS Glycoprotein VI (GPVI) is a platelet collagen/fibrin(ogen) receptor and an emerging pharmacological target for the treatment of thrombotic and thrombo-inflammatory diseases, notably ischaemic stroke. A first anti-human GPVI (hGPVI) antibody Fab-fragment (ACT017/glenzocimab, KD: 4.1 nM) recently passed a clinical phase 1b/2a study in patients with acute ischaemic stroke and was found to be well tolerated, safe, and potentially beneficial. In this study, a novel humanized anti-GPVI antibody Fab-fragment (EMA601; KD: 0.195 nM) was developed that inhibits hGPVI function with very high potency in vitro and in vivo. METHODS Fab-fragments of the mouse anti-hGPVI IgG Emf6.1 were tested for functional GPVI inhibition in human platelets and in hGPVI expressing (hGP6tg/tg) mouse platelets. The in vivo effect of Emf6.1Fab was assessed in a tail bleeding assay, an arterial thrombosis model and the transient middle cerebral artery occlusion (tMCAO) model of ischaemic stroke. Using complementary-determining region grafting, a humanized version of Emf6.1Fab (EMA601) was generated. Emf6.1Fab/EMA601 interaction with hGPVI was mapped in array format and kinetics and quantified by bio-layer interferometry. RESULTS Emf6.1Fab (KD: 0.427 nM) blocked GPVI function in human and hGP6tg/tg mouse platelets in multiple assays in vitro at concentrations ≥5 µg/mL. Emf6.1Fab (4 mg/kg)-treated hGP6tg/tg mice showed potent hGPVI inhibition ex vivo and were profoundly protected from arterial thrombosis as well as from cerebral infarct growth after tMCAO, whereas tail-bleeding times remained unaffected. Emf6.1Fab binds to a so far undescribed membrane proximal epitope in GPVI. The humanized variant EMA601 displayed further increased affinity for hGPVI (KD: 0.195 nM) and fully inhibited the receptor at 0.5 µg/mL, corresponding to a >50-fold potency compared with ACT017. CONCLUSIONS EMA601 is a conceptually novel and promising anti-platelet agent to efficiently prevent or treat arterial thrombosis and thrombo-inflammatory pathologies in humans at risk.
Collapse
Affiliation(s)
- Stefano Navarro
- Institute of Experimental Biomedicine I, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Ivan Talucci
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Str. 11, 97080 Würzburg, Germany
| | - Vanessa Göb
- Institute of Experimental Biomedicine I, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Stefanie Hartmann
- Institute of Experimental Biomedicine I, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
| | - Sarah Beck
- Institute of Experimental Biomedicine I, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | | | - Guido Stoll
- Institute of Experimental Biomedicine I, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
| | - Hans M Maric
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - David Stegner
- Institute of Experimental Biomedicine I, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine I, Josef-Schneider-Straße 2, 97080 Würzburg, Germany
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
- EMFRET Analytics GmbH, Eibelstadt, Germany
| |
Collapse
|
2
|
Fernández DI, Troitiño S, Sobota V, Tullemans BME, Zou J, van den Hurk H, García Á, Honarnejad S, Kuijpers MJE, Heemskerk JWM. Ultra-high throughput-based screening for the discovery of antiplatelet drugs affecting receptor dependent calcium signaling dynamics. Sci Rep 2024; 14:6229. [PMID: 38486006 PMCID: PMC10940705 DOI: 10.1038/s41598-024-56799-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024] Open
Abstract
Distinct platelet activation patterns are elicited by the tyrosine kinase-linked collagen receptor glycoprotein VI (GPVI) and the G-protein coupled protease-activated receptors (PAR1/4) for thrombin. This is reflected in the different platelet Ca2+ responses induced by the GPVI agonist collagen-related peptide (CRP) and the PAR1/4 agonist thrombin. Using a 96 well-plate assay with human Calcium-6-loaded platelets and a panel of 22 pharmacological inhibitors, we assessed the cytosolic Ca2+ signaling domains of these receptors and developed an automated Ca2+ curve algorithm. The algorithm was used to evaluate an ultra-high throughput (UHT) based screening of 16,635 chemically diverse small molecules with orally active physicochemical properties for effects on platelets stimulated with CRP or thrombin. Stringent agonist-specific selection criteria resulted in the identification of 151 drug-like molecules, of which three hit compounds were further characterized. The dibenzyl formamide derivative ANO61 selectively modulated thrombin-induced Ca2+ responses, whereas the aromatic sulfonyl imidazole AF299 and the phenothiazine ethopropazine affected CRP-induced responses. Platelet functional assays confirmed selectivity of these hits. Ethopropazine retained its inhibitory potential in the presence of plasma, and suppressed collagen-dependent thrombus buildup at arterial shear rate. In conclusion, targeting of platelet Ca2+ signaling dynamics in a screening campaign has the potential of identifying novel platelet-inhibiting molecules.
Collapse
Affiliation(s)
- Delia I Fernández
- The Department of Biochemistry, CARIM, Maastricht University, 6229 ER, Maastricht, The Netherlands
- Platelet Proteomics Group, CiMUS, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Sara Troitiño
- Platelet Proteomics Group, CiMUS, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Vladimír Sobota
- IHU-LIRYC, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, 33604, Bordeaux, France
- Institut de Mathématiques de Bordeaux, UMR5251, University of Bordeaux, 33 405, Talence, France
| | - Bibian M E Tullemans
- The Department of Biochemistry, CARIM, Maastricht University, 6229 ER, Maastricht, The Netherlands
- Synapse Research Institute, Kon. Emmaplein 7, 6217 KD, Maastricht, The Netherlands
| | - Jinmi Zou
- The Department of Biochemistry, CARIM, Maastricht University, 6229 ER, Maastricht, The Netherlands
- Synapse Research Institute, Kon. Emmaplein 7, 6217 KD, Maastricht, The Netherlands
| | | | - Ángel García
- Platelet Proteomics Group, CiMUS, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | | | - Marijke J E Kuijpers
- The Department of Biochemistry, CARIM, Maastricht University, 6229 ER, Maastricht, The Netherlands.
- Thrombosis Expertise Centre, Heart and Vascular Centre, Maastricht University Medical Centre+, 6229 HX, Maastricht, The Netherlands.
| | - Johan W M Heemskerk
- The Department of Biochemistry, CARIM, Maastricht University, 6229 ER, Maastricht, The Netherlands.
- Synapse Research Institute, Kon. Emmaplein 7, 6217 KD, Maastricht, The Netherlands.
| |
Collapse
|
3
|
Watanabe N, Shinozaki Y, Ogiwara S, Miyagasako R, Sasaki A, Kato J, Suzuki Y, Fukunishi N, Okada Y, Saito T, Iida Y, Higashiseto M, Masuda H, Nagata E, Gotoh K, Amino M, Tsuji T, Morita S, Nakagawa Y, Hirayama N, Inokuchi S. Diphenyl-tetrazol-propanamide Derivatives Act as Dual-Specific Antagonists of Platelet CLEC-2 and Glycoprotein VI. Thromb Haemost 2024; 124:203-222. [PMID: 37967855 DOI: 10.1055/a-2211-5202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
BACKGROUND Platelet C-type lectin-like receptor 2 (CLEC-2) induces platelet activation and aggregation after clustering by its ligand podoplanin (PDPN). PDPN, which is not normally expressed in cells in contact with blood flow, is induced in inflammatory immune cells and some malignant tumor cells, thereby increasing the risk of venous thromboembolism (VTE) and tumor metastasis. Therefore, small-molecule compounds that can interfere with the PDPN-CLEC-2 axis have the potential to become selective antiplatelet agents. METHODS AND RESULTS Using molecular docking analysis of CLEC-2 and a PDPN-CLEC-2 binding-inhibition assay, we identified a group of diphenyl-tetrazol-propanamide derivatives as novel CLEC-2 inhibitors. A total of 12 hit compounds also inhibited PDPN-induced platelet aggregation in humans and mice. Unexpectedly, these compounds also fit the collagen-binding pocket of the glycoprotein VI molecule, thereby inhibiting collagen interaction. These compounds also inhibited collagen-induced platelet aggregation, and one compound ameliorated collagen-induced thrombocytopenia in mice. For clinical use, these compounds will require a degree of chemical modification to decrease albumin binding. CONCLUSION Nonetheless, as dual activation of platelets by collagen and PDPN-positive cells is expected to occur after the rupture of atherosclerotic plaques, these dual antagonists could represent a promising pharmacophore, particularly for arterial thrombosis, in addition to VTE and metastasis.
Collapse
Affiliation(s)
- Nobuo Watanabe
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Institute of Advanced Biosciences, Tokai University, Hiratsuka, Kanagawa, Japan
| | - Yoshiko Shinozaki
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Sanae Ogiwara
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Riko Miyagasako
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Ayumi Sasaki
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Junko Kato
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Yusuke Suzuki
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Natsuko Fukunishi
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Yoshinori Okada
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Takeshi Saito
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Yumi Iida
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Misaki Higashiseto
- Support Center for Medical Research and Education, Tokai University, Isehara, Kanagawa, Japan
| | - Haruchika Masuda
- Department of Physiology, Tokai University School of Medicine, Shimokasuya, Isehara, Kanagawa, Japan
| | - Eiichiro Nagata
- Department of Neurology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Kazuhito Gotoh
- Department of Laboratory Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Mari Amino
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Tomoatsu Tsuji
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Seiji Morita
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Yoshihide Nakagawa
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Noriaki Hirayama
- Institute of Advanced Biosciences, Tokai University, Hiratsuka, Kanagawa, Japan
- The Institute of Medical Sciences, Tokai University, Isehara, Kanagawa, Japan
| | - Sadaki Inokuchi
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Institute of Advanced Biosciences, Tokai University, Hiratsuka, Kanagawa, Japan
| |
Collapse
|
4
|
Haffouz A, Elleuch H, Khemakhem B, Ben Amor I, Jerbi A, Gargouri J, Sahli E, Mhadhbi N, Ghalla H, Rezgui F, Gargouri A, HadjKacem B. Antiplatelet activity and toxicity profile of novel phosphonium salts derived from Michael reaction. Eur J Pharm Sci 2024; 194:106692. [PMID: 38181870 DOI: 10.1016/j.ejps.2024.106692] [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: 10/03/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/07/2024]
Abstract
In this work, five novel phosphonium salts derived from the Michael reaction were screened for their antiplatelet activity. Our findings revealed that compounds 2a, 2b, 2c, and 2d significantly inhibit platelet aggregation triggered by ADP or collagen (P < 0.001). Notably, compound 2c inhibited the arachidonic acid pathway (P < 0.001). Moreover, the selected compounds reduce CD62-P expression and inhibit GPIIb/IIIa activation. The interactions of the active compounds with their targets, ADP and collagen receptors, P2Y12 and GPVI respectively were investigated in silico using molecular docking studies. The results revealed a strong affinity of the active compounds for P2Y12 and GPVI. Additionally, cytotoxicity assays on platelets, erythrocytes, and human embryonic kidney HEK293 cells showed that compounds 2a, 2c and 2d were non-toxic even at high concentrations. In summary, our study shows that phosphonium salts can have strong antiplatelet power and suggests that compounds 2a, 2c and 2d could be promising antiplatelet agents for the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Asma Haffouz
- Laboratory of Molecular Biotechnology of Eucaryotes, Centre of Biotechnology of Sfax, University of Sfax, B.P 1177, 3018, Sfax, Tunisia
| | - Haitham Elleuch
- Laboratory of Organic Chemistry, Faculty of Sciences, University Campus, 2092, University of Tunis El Manar, Tunis, Tunisia
| | - Bassem Khemakhem
- Laboratory of Plant Biotechnology, Sfax Faculty of Sciences, BP 1171, University of Sfax, 3038 Sfax, Tunisia
| | - Ikram Ben Amor
- Laboratory of Hematology (LR19SP04), Medical Faculty of Sfax. University of Sfax, Magida Boulila Avenue, 3029 Sfax, Tunisia
| | - Amira Jerbi
- Laboratory of Hematology (LR19SP04), Medical Faculty of Sfax. University of Sfax, Magida Boulila Avenue, 3029 Sfax, Tunisia
| | - Jalel Gargouri
- Laboratory of Hematology (LR19SP04), Medical Faculty of Sfax. University of Sfax, Magida Boulila Avenue, 3029 Sfax, Tunisia
| | - Emna Sahli
- Analytical service provider unit, Centre of Biotechnology of Sfax, University of Sfax, 3018, Sfax, Tunisia
| | - Noureddine Mhadhbi
- Laboratory Physico Chemistry of the Solid State, Department of Chemistry, Faculty of Sciences, University of Sfax, BP 1171, 3000 Sfax, Tunisia; University of Monastir, Preparatory Institute for Engineering Studies of Monastir, 5019 Monastir, Tunisia
| | - Houcine Ghalla
- Quantum Physics and Statistic Laboratory, Faculty of Sciences, University of Monastir, Monastir, 5000, Tunisia
| | - Farhat Rezgui
- Laboratory of Organic Chemistry, Faculty of Sciences, University Campus, 2092, University of Tunis El Manar, Tunis, Tunisia
| | - Ali Gargouri
- Laboratory of Molecular Biotechnology of Eucaryotes, Centre of Biotechnology of Sfax, University of Sfax, B.P 1177, 3018, Sfax, Tunisia
| | - Basma HadjKacem
- Laboratory of Molecular Biotechnology of Eucaryotes, Centre of Biotechnology of Sfax, University of Sfax, B.P 1177, 3018, Sfax, Tunisia; Department of Life Sciences, Faculty of Sciences of Gafsa, University of Gafsa, Gafsa, Tunisia.
| |
Collapse
|
5
|
Foster H, Wilson C, Gauer JS, Xu RG, Howard MJ, Manfield IW, Ariëns R, Naseem K, Vidler LR, Philippou H, Foster R. A Comparative Assessment Study of Known Small-molecule GPVI Modulators. ACS Med Chem Lett 2022; 13:171-181. [PMID: 35178172 PMCID: PMC8842102 DOI: 10.1021/acsmedchemlett.1c00414] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/10/2021] [Indexed: 12/11/2022] Open
Abstract
The GPVI platelet receptor was recently validated as a safe antiplatelet target for the treatment of thrombosis using several peptidic modulators. In contrast, few weakly potent small-molecule GPVI antagonists have been reported. Those that have been published often lack evidence for target engagement, and their biological efficacy cannot be compared because of the natural donor variability associated with the assays implemented. Herein, we present the first side-by-side assessment of the reported GPVI small-molecule modulators. We have characterized their functional activities on platelet activation and aggregation using flow cytometry as well as light transmission and electrical impedance aggregometry. We also utilized microscale thermophoresis (MST) and saturation transfer difference (STD) NMR to validate GPVI binding and have used this along with molecular modeling to suggest potential binding interactions. We conclude that of the compounds examined, losartan and compound 5 are currently the most viable GPVI modulators.
Collapse
Affiliation(s)
- Holly Foster
- School
of Chemistry, University of Leeds, Leeds LS2 9JT, U.K.
- Leeds
Institute of Cardiovascular and Metabolic Medicine (LICAMM), School
of Medicine, University of Leeds, Leeds LS2 9JT, U.K.
| | - Clare Wilson
- Leeds
Institute of Cardiovascular and Metabolic Medicine (LICAMM), School
of Medicine, University of Leeds, Leeds LS2 9JT, U.K.
| | - Julia S. Gauer
- Leeds
Institute of Cardiovascular and Metabolic Medicine (LICAMM), School
of Medicine, University of Leeds, Leeds LS2 9JT, U.K.
| | - Rui-Gang Xu
- Leeds
Institute of Cardiovascular and Metabolic Medicine (LICAMM), School
of Medicine, University of Leeds, Leeds LS2 9JT, U.K.
| | - Mark J. Howard
- School
of Chemistry, University of Leeds, Leeds LS2 9JT, U.K.
| | - Iain W. Manfield
- Faculty
of Biological Sciences and Astbury Centre for Structural Molecular
Biology, University of Leeds, Leeds LS2 9JT, U.K.
| | - Robert Ariëns
- Leeds
Institute of Cardiovascular and Metabolic Medicine (LICAMM), School
of Medicine, University of Leeds, Leeds LS2 9JT, U.K.
| | - Khalid Naseem
- Leeds
Institute of Cardiovascular and Metabolic Medicine (LICAMM), School
of Medicine, University of Leeds, Leeds LS2 9JT, U.K.
| | | | - Helen Philippou
- Leeds
Institute of Cardiovascular and Metabolic Medicine (LICAMM), School
of Medicine, University of Leeds, Leeds LS2 9JT, U.K.
| | - Richard Foster
- School
of Chemistry, University of Leeds, Leeds LS2 9JT, U.K.
- Leeds
Institute of Cardiovascular and Metabolic Medicine (LICAMM), School
of Medicine, University of Leeds, Leeds LS2 9JT, U.K.
| |
Collapse
|
6
|
Olğaç S, Olğaç A, Yenicesu İ, Özkan Y. Identification of Novel Antiplatelet Agents by Targeting Glycoprotein VI: A Combined Virtual Screening Study. Bioorg Chem 2022; 121:105661. [DOI: 10.1016/j.bioorg.2022.105661] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 01/18/2022] [Accepted: 02/05/2022] [Indexed: 11/28/2022]
|
7
|
Wu Y, Zeng Z, Guo Y, Song L, Weatherhead JE, Huang X, Zeng Y, Bimler L, Chang CY, Knight JM, Valladolid C, Sun H, Cruz MA, Hube B, Naglik JR, Luong AU, Kheradmand F, Corry DB. Candida albicans elicits protective allergic responses via platelet mediated T helper 2 and T helper 17 cell polarization. Immunity 2021; 54:2595-2610.e7. [PMID: 34506733 DOI: 10.1016/j.immuni.2021.08.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/19/2021] [Accepted: 08/10/2021] [Indexed: 12/24/2022]
Abstract
Fungal airway infection (airway mycosis) is an important cause of allergic airway diseases such as asthma, but the mechanisms by which fungi trigger asthmatic reactions are poorly understood. Here, we leverage wild-type and mutant Candida albicans to determine how this common fungus elicits characteristic Th2 and Th17 cell-dependent allergic airway disease in mice. We demonstrate that rather than proteinases that are essential virulence factors for molds, C. albicans instead promoted allergic airway disease through the peptide toxin candidalysin. Candidalysin activated platelets through the Von Willebrand factor (VWF) receptor GP1bα to release the Wnt antagonist Dickkopf-1 (Dkk-1) to drive Th2 and Th17 cell responses that correlated with reduced lung fungal burdens. Platelets simultaneously precluded lethal pulmonary hemorrhage resulting from fungal lung invasion. Thus, in addition to hemostasis, platelets promoted protection against C. albicans airway mycosis through an antifungal pathway involving candidalysin, GP1bα, and Dkk-1 that promotes Th2 and Th17 responses.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Pathology & Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Zhimin Zeng
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Yubiao Guo
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Lizhen Song
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jill E Weatherhead
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; The National School of Tropical Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Xinyan Huang
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yuying Zeng
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Lynn Bimler
- Department of Pathology & Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; The National School of Tropical Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Cheng-Yen Chang
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; The Translational Biology and Molecular Medicine Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - John M Knight
- Department of Pathology & Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; The Biology of Inflammation Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Christian Valladolid
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular Physiology & Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Michael E. DeBakey VA Center for Translational Research on Inflammatory Diseases, Houston Texas, 77030, USA
| | - Hua Sun
- Department of Otolaryngology, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Miguel A Cruz
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Michael E. DeBakey VA Center for Translational Research on Inflammatory Diseases, Houston Texas, 77030, USA
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute Jena (HKI), Jena 07745, Germany; Institute of Microbiology, Friedrich Schiller University, Jena 07737, Germany
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 1UL, UK
| | - Amber U Luong
- Department of Otolaryngology, McGovern Medical School of the University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Farrah Kheradmand
- Department of Pathology & Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; The Biology of Inflammation Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Michael E. DeBakey VA Center for Translational Research on Inflammatory Diseases, Houston Texas, 77030, USA
| | - David B Corry
- Department of Pathology & Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; The Biology of Inflammation Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Michael E. DeBakey VA Center for Translational Research on Inflammatory Diseases, Houston Texas, 77030, USA.
| |
Collapse
|
8
|
Jiménez-Orozco FA, Galicia-Zapatero S, López-López E, Medina-Franco JL, Cedeño FL, Flores-García M, Mejia-Domínguez A, de la Peña-Díaz A. Monosubstituted Coumarins Inhibit Epinephrine-Induced Platelet Aggregation Antiplatelet Effect of Monosubstituted Coumarins. Cardiovasc Hematol Agents Med Chem 2021; 20:43-51. [PMID: 33906594 PMCID: PMC9127734 DOI: 10.2174/1871525719666210427132808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 03/15/2021] [Accepted: 03/29/2021] [Indexed: 11/22/2022]
Abstract
Aim The aim of this study was to evaluate the in vitro effect of coumarin and 15 monosubstituted derivatives on the inhibition of human platelet aggregation induced by various pro-aggregatory agonists, particularly by epinephrine. Background The emergence of residual platelet reactivity during the use of conventional antiplatelet agents (acetylsalicylic acid and clopidogrel) is one of the main causes of double therapy´s therapeutic failure. Platelet adrenoceptors participate in residual platelet reactivity. Therefore, it is necessary to develop new antiplatelet agents that inhibit epinephrine-induced platelet aggregation as a new therapeutic strategy. Information on the antiplatelet activity of coumarins in inhibiting epinephrine-induced aggregation is limited. Objective The objective of this study was to establish the structure-activity relationship (SAR) of coumarin derivatives with hydroxy, methoxy, and acetoxy groups in different positions of the coumarin nucleus to identify the most active molecules. Moreover, this study aimed to use in silico studies to suggest potential drug targets to which the molecules bind to produce antiplatelet effects. Methods The platelet aggregation was performed using a Lumi-aggregometer; the inhibitory activity of 16 compounds were evaluated by inducing the aggregation of human platelets (250 × 103/μl) with epinephrine (10 µM), collagen (2 µg/ml) or ADP (10 µM). The aggregation of control platelets was considered 100% of the response for each pro-aggregatory agonist. Results Eleven molecules inhibited epinephrine-induced aggregation, with 3-acetoxycoumarin and 7-methoxycoumarin being the most active. Only coumarin inhibited collagen-induced platelet aggregation, but no molecule showed activity when using ADP as an inducer. Conclusions In silico studies suggest that most active molecules might have antagonistic interactions in the α2 and β2 adrenoceptors. The antiplatelet actions of these coumarins have the potential to reduce residual platelet reactivity and thus contribute to the development of future treatments for patients who do not respond adequately to conventional agents.
Collapse
Affiliation(s)
- Fausto Alejandro Jiménez-Orozco
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán 04510, CDMX. Mexico
| | - Sergio Galicia-Zapatero
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán 04510, CDMX. Mexico
| | - Edgar López-López
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán 04510, CDMX. Mexico
| | - José L Medina-Franco
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán 04510, CDMX. Mexico
| | - Fernando León Cedeño
- Departamento de Química Orgánica, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán 04510, CDMX. Mexico
| | - Mirthala Flores-García
- Departamento de Biología Molecular, Instituto Nacional de Cardiología Ignacio Chávez, Tlalpan 14080, CDMX. Mexico
| | - Ana Mejia-Domínguez
- Banco de Sangre, Instituto Nacional de Cardiología Ignacio Chávez, Tlalpan 14080, CDMX. Mexico
| | - Aurora de la Peña-Díaz
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán 04510, CDMX. Mexico
| |
Collapse
|
9
|
Damaskinaki FN, Moran LA, Garcia A, Kellam B, Watson SP. Overcoming challenges in developing small molecule inhibitors for GPVI and CLEC-2. Platelets 2021; 32:744-752. [PMID: 33406951 DOI: 10.1080/09537104.2020.1863939] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
GPVI and CLEC-2 have emerged as promising targets for long-term prevention of both arterial thrombosis and thrombo-inflammation with a decreased bleeding risk relative to current drugs. However, while there are potent blocking antibodies of both receptors, their protein nature comes with decreased bioavailability, making formulation for oral medication challenging. Small molecules are able to overcome these limitations, but there are many challenges in developing antagonists of nanomolar potency, which is necessary when considering the structural features that underlie the interaction of CLEC-2 and GPVI with their protein ligands. In this review, we describe current small-molecule inhibitors for both receptors and strategies to overcome such limitations, including considerations when it comes to in silico drug design and the importance of complex compound library selection.
Collapse
Affiliation(s)
- Foteini-Nafsika Damaskinaki
- Institute of Cardiovascular Sciences, Level 1 IBR, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), The Universities of Birmingham and Nottingham, The Midlands, UK.,Biodiscovery Institute, University Park, University of Nottingham, Nottingham, UK
| | - Luis A Moran
- Institute of Cardiovascular Sciences, Level 1 IBR, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, and Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Angel Garcia
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, and Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Barrie Kellam
- Centre of Membrane Proteins and Receptors (COMPARE), The Universities of Birmingham and Nottingham, The Midlands, UK.,Biodiscovery Institute, University Park, University of Nottingham, Nottingham, UK
| | - Steve P Watson
- Institute of Cardiovascular Sciences, Level 1 IBR, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), The Universities of Birmingham and Nottingham, The Midlands, UK
| |
Collapse
|
10
|
Foster H, Wilson C, Philippou H, Foster R. Progress toward a Glycoprotein VI Modulator for the Treatment of Thrombosis. J Med Chem 2020; 63:12213-12242. [PMID: 32463237 DOI: 10.1021/acs.jmedchem.0c00262] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Pathogenic thrombus formation accounts for the etiology of many serious conditions including myocardial infarction, stroke, deep vein thrombosis, and pulmonary embolism. Despite the development of numerous anticoagulants and antiplatelet agents, the mortality rate associated with these diseases remains high. In recent years, however, significant epidemiological evidence and clinical models have emerged to suggest that modulation of the glycoprotein VI (GPVI) platelet receptor could be harnessed as a novel antiplatelet strategy. As such, many peptidic agents have been described in the past decade, while more recent efforts have focused on the development of small molecule modulators. Herein the rationale for targeting GPVI is summarized and the published GPVI modulators are reviewed, with particular focus on small molecules. A qualitative pharmacophore hypothesis for small molecule ligands at GPVI is also presented.
Collapse
Affiliation(s)
- Holly Foster
- School of Chemistry and Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds LS2 9JT, U.K
| | - Clare Wilson
- Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds LS2 9JT, U.K
| | - Helen Philippou
- Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds LS2 9JT, U.K
| | - Richard Foster
- School of Chemistry and Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), School of Medicine, University of Leeds, Leeds LS2 9JT, U.K
| |
Collapse
|
11
|
Durrant TN, Hers I. PI3K inhibitors in thrombosis and cardiovascular disease. Clin Transl Med 2020; 9:8. [PMID: 32002690 PMCID: PMC6992830 DOI: 10.1186/s40169-020-0261-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/13/2020] [Indexed: 12/15/2022] Open
Abstract
Phosphoinositide 3-kinases (PI3Ks) are lipid kinases that regulate important intracellular signalling and vesicle trafficking events via the generation of 3-phosphoinositides. Comprising eight core isoforms across three classes, the PI3K family displays broad expression and function throughout mammalian tissues, and the (patho)physiological roles of these enzymes in the cardiovascular system present the PI3Ks as potential therapeutic targets in settings such as thrombosis, atherosclerosis and heart failure. This review will discuss the PI3K enzymes and their roles in cardiovascular physiology and disease, with a particular focus on platelet function and thrombosis. The current progress and future potential of targeting the PI3K enzymes for therapeutic benefit in cardiovascular disease will be considered, while the challenges of developing drugs against these master cellular regulators will be discussed.
Collapse
Affiliation(s)
- Tom N Durrant
- Department of Chemistry, University of Oxford, Oxford, OX1 3QZ, UK.
| | - Ingeborg Hers
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.
| |
Collapse
|
12
|
Gaspar RS, da Silva SA, Stapleton J, Fontelles JLDL, Sousa HR, Chagas VT, Alsufyani S, Trostchansky A, Gibbins JM, Paes AMDA. Myricetin, the Main Flavonoid in Syzygium cumini Leaf, Is a Novel Inhibitor of Platelet Thiol Isomerases PDI and ERp5. Front Pharmacol 2020; 10:1678. [PMID: 32116678 PMCID: PMC7011086 DOI: 10.3389/fphar.2019.01678] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/23/2019] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Flavonoids have been characterized as a prominent class of compounds to treat thrombotic diseases through the inhibition of thiol isomerases. Syzygium cumini is a flavonoid-rich medicinal plant that contains myricetin and gallic acid. Little is known about the potential antiplatelet properties of S. cumini and its constituent flavonoids. OBJECTIVE To evaluate the antiplatelet effects and mechanism of action of a polyphenol-rich extract (PESc) from S. cumini leaf and its most prevalent polyphenols, myricetin and gallic acid. METHODS PESc, myricetin, and gallic acid were incubated with platelet-rich plasma and washed platelets to assess platelet aggregation and activation. In vitro platelet adhesion and thrombus formation as well as in vivo bleeding time were performed. Finally, myricetin was incubated with recombinant thiol isomerases to assess its potential to bind and inhibit these, while molecular docking studies predicted possible binding sites. RESULTS PESc decreased platelet activation and aggregation induced by different agonists. Myricetin exerted potent antiplatelet effects, whereas gallic acid did not. Myricetin reduced the ability of platelets to spread on collagen, form thrombi in vitro without affecting hemostasis in vivo. Fluorescence quenching studies suggested myricetin binds to different thiol isomerases with similar affinity, despite inhibiting only protein disulfide isomerase (PDI) and ERp5 reductase activities. Finally, molecular docking studies suggested myricetin formed non-covalent bonds with PDI and ERp5. CONCLUSIONS PESc and its most abundant flavonoid myricetin strongly inhibit platelet function. Additionally, myricetin is a novel inhibitor of ERp5 and PDI, unveiling a new therapeutic perspective for the treatment of thrombotic disorders.
Collapse
Affiliation(s)
- Renato Simões Gaspar
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, Brazil
| | - Samira Abdalla da Silva
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, Brazil
| | - Jennifer Stapleton
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - João Lucas de Lima Fontelles
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, Brazil
| | - Hiran Reis Sousa
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, Brazil
| | - Vinicyus Teles Chagas
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, Brazil
| | - Shuruq Alsufyani
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Andrés Trostchansky
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Jonathan M. Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Antonio Marcus de Andrade Paes
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, São Luís, Brazil
| |
Collapse
|
13
|
Cammisotto V, Carnevale R, Nocella C, Stefanini L, Bartimoccia S, Coluccia A, Silvestri R, Pignatelli P, Pastori D, Violi F. Nox2-mediated platelet activation by glycoprotein (GP) VI: Effect of rivaroxaban alone and in combination with aspirin. Biochem Pharmacol 2019; 163:111-118. [DOI: 10.1016/j.bcp.2019.02.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 02/12/2019] [Indexed: 01/04/2023]
|
14
|
Onselaer MB, Nagy M, Pallini C, Pike JA, Perrella G, Quintanilla LG, Eble JA, Poulter NS, Heemskerk JWM, Watson SP. Comparison of the GPVI inhibitors losartan and honokiol. Platelets 2019; 31:187-197. [PMID: 30849265 PMCID: PMC7034533 DOI: 10.1080/09537104.2019.1585526] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Losartan and honokiol are small molecules which have been described to inhibit aggregation of platelets by collagen. Losartan has been proposed to block clustering of GPVI but not to affect binding of collagen. Honokiol has been reported to bind directly to GPVI but only at a concentration that is three orders of magnitude higher than that needed for inhibition of aggregation. The mechanism of action of both inhibitors is so far unclear. In the present study, we confirm the inhibitory effects of both agents on platelet aggregation by collagen and show that both also block the aggregation induced by the activation of CLEC-2 or the low affinity immune receptor FcγRIIa at similar concentrations. For GPVI and CLEC-2, this inhibition is associated with a reduction in protein tyrosine phosphorylation of multiple proteins including Syk. In contrast, on a collagen surface, spreading of platelets and clustering of GPVI (measured by single molecule localisation microscopy) was not altered by losartan or honokiol. Furthermore, in flow whole-blood, both inhibitors suppressed the formation of multi-layered platelet thrombi at arteriolar shear rates at concentrations that hardly affect collagen-induced platelet aggregation in platelet rich plasma. Together, these results demonstrate that losartan and honokiol have multiple effects on platelets which should be considered in the use of these compounds as anti-platelet agents.
Collapse
Affiliation(s)
- Marie-Blanche Onselaer
- Institute of Cardiovascular Sciences, IBR Building, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Magdolna Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, MD, The Netherlands
| | - Chiara Pallini
- Institute of Cardiovascular Sciences, IBR Building, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Jeremy A Pike
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands
| | - Gina Perrella
- Institute of Cardiovascular Sciences, IBR Building, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, MD, The Netherlands
| | - Lourdes Garcia Quintanilla
- Institute of Cardiovascular Sciences, IBR Building, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Natalie S Poulter
- Institute of Cardiovascular Sciences, IBR Building, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, MD, The Netherlands
| | - Steve P Watson
- Institute of Cardiovascular Sciences, IBR Building, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands
| |
Collapse
|
15
|
Martins Lima A, Bragina ME, Burri O, Bortoli Chapalay J, Costa-Fraga FP, Chambon M, Fraga-Silva RA, Stergiopulos N. An optimized and validated 384-well plate assay to test platelet function in a high-throughput screening format. Platelets 2018; 30:563-571. [PMID: 30183501 DOI: 10.1080/09537104.2018.1514106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Despite significant advances in the treatment of cardiovascular diseases, antiplatelet therapies are still associated with a high risk of hemorrhage. In order to develop new drugs, methods to measure platelet function must be adapted for the high-throughput screening (HTS) format. Currently, all assays capable of assessing platelet function are either expensive, complex, or not validated, which makes them unsuitable for drug discovery. Here, we propose a simple, low-cost, and high-throughput-compatible platelet function assay, validated for the 384-well plate. In the proposed assay, agonist-induced platelet activity was assessed by three different methods: (i) measurement of light absorbance, which decreases with platelet aggregation; (ii) luminescence measurement, based on ATP release from activated platelets and luciferin-luciferase reaction; and (iii) automated bright-field microscopy of the wells and further quantification of platelet image area, described here for the first time. Brightfield imaging results were validated by demonstrating the similarity of dose-response curves obtained with absorbance and luminescence measurements after stimulating platelets, pre-incubated with prostaglandin E1 or tirofiban, and demonstrating the similarity of dose-response curves obtained with agonists. Assay quality was confirmed using the Z'-factor, a statistical parameter used to validate the robustness and suitability of an HTS assay. The results showed that, under high rotations per minute (1200 RPM), an acceptable Z'-factor score is reached for absorbance measurements (Z'-factor - 0.58) and automated brightfield imaging (Z'-factor - 0.52), without the need of replicates, while triplicates must be used to achieve an acceptable Z'-factor score (0.54) for luminescence measurements. Using low platelet concentration (4 × 104/μl - 10 μl), the brightfield imaging test was further validated using washed platelets. Furthermore, drug screening was performed with compounds selected by structure-based virtual screening. Taken together, this study presents an optimized and validated assay for HTS to be used as a tool for antiplatelet drug discovery.
Collapse
Affiliation(s)
- Augusto Martins Lima
- a Institute of Bioengineering , École Polytechnique Fédérale de Lausanne , Lausanne , Switzerland
| | - Maiia E Bragina
- a Institute of Bioengineering , École Polytechnique Fédérale de Lausanne , Lausanne , Switzerland
| | - Olivier Burri
- b BioImaging and Optics Core Facility , École Polytechnique Fédérale de Lausanne , Lausanne , Switzerland
| | - Julien Bortoli Chapalay
- c Biomolecular Screening Facility , École Polytechnique Federale de Lausanne , Lausanne , Switzerland
| | - Fabiana P Costa-Fraga
- a Institute of Bioengineering , École Polytechnique Fédérale de Lausanne , Lausanne , Switzerland
| | - Marc Chambon
- c Biomolecular Screening Facility , École Polytechnique Federale de Lausanne , Lausanne , Switzerland
| | - Rodrigo A Fraga-Silva
- a Institute of Bioengineering , École Polytechnique Fédérale de Lausanne , Lausanne , Switzerland
| | - Nikolaos Stergiopulos
- a Institute of Bioengineering , École Polytechnique Fédérale de Lausanne , Lausanne , Switzerland
| |
Collapse
|
16
|
Elaskalani O, Abdol Razak NB, Metharom P. Neutrophil extracellular traps induce aggregation of washed human platelets independently of extracellular DNA and histones. Cell Commun Signal 2018; 16:24. [PMID: 29843771 PMCID: PMC5975482 DOI: 10.1186/s12964-018-0235-0] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/15/2018] [Indexed: 11/13/2022] Open
Abstract
Background The release of neutrophil extracellular traps (NETs), a mesh of DNA, histones and neutrophil proteases from neutrophils, was first demonstrated as a host defence against pathogens. Recently it became clear that NETs are also released in pathological conditions. NETs released in the blood can activate thrombosis and initiate a cascade of platelet responses. However, it is not well understood if these responses are mediated through direct or indirect interactions. We investigated whether cell-free NETs can induce aggregation of washed human platelets in vitro and the contribution of NET-derived extracellular DNA and histones to platelet activation response. Methods Isolated human neutrophils were stimulated with PMA to produce robust and consistent NETs. Cell-free NETs were isolated and characterised by examining DNA-histone complexes and quantification of neutrophil elastase with ELISA. NETs were incubated with washed human platelets to assess several platelet activation responses. Using pharmacological inhibitors, we explored the role of different NET components, as well as main platelet receptors, and downstream signalling pathways involved in NET-induced platelet aggregation. Results Cell-free NETs directly induced dose-dependent platelet aggregation, dense granule secretion and procoagulant phosphatidyl serine exposure on platelets. Surprisingly, we found that inhibition of NET-derived DNA and histones did not affect NET-induced platelet aggregation or activation. We further identified the molecular pathways involved in NET-activated platelets. The most potent single modulator of NET-induced platelet responses included NET-bound cathepsin G, platelet Syk kinase, and P2Y12 and αIIbβ3 receptors. Conclusions In vitro-generated NETs can directly induce marked aggregation of washed human platelets. Pre-treatment of NETs with DNase or heparin did not reduce NET-induced activation or aggregation of human washed platelets. We further identified the molecular pathways activated in platelets in response to NETs. Taken together, we conclude that targeting certain platelet activation pathways, rather than the NET scaffold, has a more profound reduction on NET-induced platelet aggregation. Electronic supplementary material The online version of this article (10.1186/s12964-018-0235-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Omar Elaskalani
- Platelet Research Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley Campus, Office 160, Building 305, Kent Street, Bentley, Perth, WA, 6102, Australia
| | - Norbaini Binti Abdol Razak
- Platelet Research Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley Campus, Office 160, Building 305, Kent Street, Bentley, Perth, WA, 6102, Australia
| | - Pat Metharom
- Platelet Research Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health and Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley Campus, Office 160, Building 305, Kent Street, Bentley, Perth, WA, 6102, Australia.
| |
Collapse
|
17
|
Elaskalani O, Khan I, Morici M, Matthysen C, Sabale M, Martins RN, Verdile G, Metharom P. Oligomeric and fibrillar amyloid beta 42 induce platelet aggregation partially through GPVI. Platelets 2017; 29:415-420. [PMID: 29206067 DOI: 10.1080/09537104.2017.1401057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The effects of the Alzheimer's disease (AD)-associated Amyloid-β (Aβ) peptides on platelet aggregation have been previously assessed, but most of these studies focused on Aβ40 species. It also remains to be determined which distinct forms of Aβ peptides exert differential effects on platelets. In AD, oligomeric Aβ42 species is widely thought to be a major contributor to the disease pathogenesis. We, therefore, examine the ability of oligomeric and fibrillary Aβ42 to affect platelet aggregation. We show that both forms of Aβ42 induced significant platelet aggregation and that it is a novel ligand for the platelet receptor GPVI. Furthermore, a novel binding peptide that reduces the formation of soluble Aβ42 oligomers was effective at preventing Aβ42-dependent platelet aggregation. These results support a role for Aβ42 oligomers in platelet hyperactivity.
Collapse
Affiliation(s)
- O Elaskalani
- a School of Biomedical Sciences, Faculty of Health Sciences , Curtin Health Innovation Research Institute, Curtin University , Perth , Australia
| | - I Khan
- a School of Biomedical Sciences, Faculty of Health Sciences , Curtin Health Innovation Research Institute, Curtin University , Perth , Australia
| | - M Morici
- b School of Medical Sciences , Centre of Excellence for Alzheimer's Disease Research and Care, Edith Cowan University , Joondalup , WA , Australia
| | - C Matthysen
- a School of Biomedical Sciences, Faculty of Health Sciences , Curtin Health Innovation Research Institute, Curtin University , Perth , Australia
| | - M Sabale
- a School of Biomedical Sciences, Faculty of Health Sciences , Curtin Health Innovation Research Institute, Curtin University , Perth , Australia
| | - R N Martins
- a School of Biomedical Sciences, Faculty of Health Sciences , Curtin Health Innovation Research Institute, Curtin University , Perth , Australia.,c Department of Biomedical Sciences, Faculty of Medicine and Health Sciences , Macquarie University , NSW , Sydney , Australia
| | - G Verdile
- a School of Biomedical Sciences, Faculty of Health Sciences , Curtin Health Innovation Research Institute, Curtin University , Perth , Australia.,b School of Medical Sciences , Centre of Excellence for Alzheimer's Disease Research and Care, Edith Cowan University , Joondalup , WA , Australia.,d School of Psychiatry and Clinical Neurosciences , University of WA , Perth , Australia
| | - P Metharom
- e Faculty of Health Sciences , Curtin Health Innovation Research Institute, Curtin University , Perth , Australia
| |
Collapse
|
18
|
Bhunia SS, Saxena AK. Molecular modelling studies in explaining the higher GPVI antagonistic activity of the racemic 2-(4-methoxyphenylsulfonyl)-2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indole-3-carboxamide than its enantiomers. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2017; 28:783-799. [PMID: 29135287 DOI: 10.1080/1062936x.2017.1396247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 10/20/2017] [Indexed: 06/07/2023]
Abstract
The GPVI receptor on the platelets plays a major role in inhibiting arterial thrombosis with limited risk of bleeding and is considered a potential anti-thrombotic target for arterial thrombosis. In the reported anti-thrombotics, tetrahydropyridoindoles, the title compound was the best inhibitor of the collagen mediated platelet aggregation by antagonizing the platelet receptor GPVI. Interestingly, the racemic title compound showed better antagonism (IC50 racemate = 6.7 μM) than either of its enantiomers (IC50 S enantiomer = 25.3 μM; IC50 R enantiomer = 126.3 μM). In order to explain this, the molecular modelling approaches viz. site map analysis, protein-protein docking and molecular dynamics simulation were carried out, which led to the identification of a second binding site located near the primary antagonist binding site known to bind losartan. The induced fit docking studies for both the enantiomers at the primary and secondary binding sites showed that the S-enantiomer has better interactions at the primary binding site than the R-enantiomer, while the R-enantiomer has better interactions at the secondary site than the S-enantiomer. Hence, the overall interactions of the racemic compound containing equimolar mixture may be higher than any one of the enantiomers and may explain the higher activity than its enantiomers of the racemic compound.
Collapse
Affiliation(s)
- S S Bhunia
- a Division of Medicinal and Process Chemistry , CSIR-Central Drug Research Institute , Lucknow , India
| | - A K Saxena
- a Division of Medicinal and Process Chemistry , CSIR-Central Drug Research Institute , Lucknow , India
| |
Collapse
|
19
|
Arthur JF, Jandeleit-Dahm K, Andrews RK. Platelet Hyperreactivity in Diabetes: Focus on GPVI Signaling-Are Useful Drugs Already Available? Diabetes 2017; 66:7-13. [PMID: 27999100 DOI: 10.2337/db16-1098] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 10/24/2016] [Indexed: 11/13/2022]
Abstract
Adults with diabetes are 2-4 times more likely to suffer from heart disease or ischemic stroke than adults without diabetes, yet standard antiplatelet therapy, which is the cornerstone for primary and secondary prevention of cardiovascular disease, fails in many patients with diabetes. Three independent but often interrelated variables that contribute to platelet hyperreactivity-high blood glucose, oxidative stress, and elevated vascular shear forces-coexist in patients with diabetes, creating a perilous concurrence of risk factors for cardiovascular events. Recent research has focused attention on the platelet-specific collagen receptor glycoprotein VI (GPVI) as a potential antithrombotic target. Signaling events downstream of GPVI are influenced by hyperglycemia, oxidative stress, and shear stress. Importantly, drugs targeting these GPVI signaling pathways are already in existence. The potential to repurpose existing drugs is a high-gain strategy for yielding new antiplatelet agents and could have particular benefit in individuals with diabetes.
Collapse
Affiliation(s)
- Jane F Arthur
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| | | | - Robert K Andrews
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| |
Collapse
|
20
|
Bhunia SS, Misra A, Khan IA, Gaur S, Jain M, Singh S, Saxena A, Hohlfield T, Dikshit M, Saxena AK. Novel Glycoprotein VI Antagonists as Antithrombotics: Synthesis, Biological Evaluation, and Molecular Modeling Studies on 2,3-Disubstituted Tetrahydropyrido(3,4-b)indoles. J Med Chem 2016; 60:322-337. [DOI: 10.1021/acs.jmedchem.6b01360] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Shome S. Bhunia
- Academy of Scientific and Innovative Research, New Delhi 110 025, India
| | | | | | | | | | | | - Aaruni Saxena
- Institut
für Pharmakologie und Klinische Pharmakologie, Heinrich-Heine-Universität Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Thomas Hohlfield
- Institut
für Pharmakologie und Klinische Pharmakologie, Heinrich-Heine-Universität Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | | | - Anil K. Saxena
- Academy of Scientific and Innovative Research, New Delhi 110 025, India
| |
Collapse
|
21
|
Snell TW, Johnston RK, Srinivasan B, Zhou H, Gao M, Skolnick J. Repurposing FDA-approved drugs for anti-aging therapies. Biogerontology 2016; 17:907-920. [PMID: 27484416 PMCID: PMC5065615 DOI: 10.1007/s10522-016-9660-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 07/25/2016] [Indexed: 12/31/2022]
Abstract
There is great interest in drugs that are capable of modulating multiple aging pathways, thereby delaying the onset and progression of aging. Effective strategies for drug development include the repurposing of existing drugs already approved by the FDA for human therapy. FDA approved drugs have known mechanisms of action and have been thoroughly screened for safety. Although there has been extensive scientific activity in repurposing drugs for disease therapy, there has been little testing of these drugs for their effects on aging. The pool of FDA approved drugs therefore represents a large reservoir of drug candidates with substantial potential for anti-aging therapy. In this paper we employ FINDSITEcomb, a powerful ligand homology modeling program, to identify binding partners for proteins produced by temperature sensing genes that have been implicated in aging. This list of drugs with potential to modulate aging rates was then tested experimentally for lifespan and healthspan extension using a small invertebrate model. Three protein targets of the rotifer Brachionus manjavacas corresponding to products of the transient receptor potential gene 7, ribosomal protein S6 polypeptide 2 gene, or forkhead box C gene, were screened against a compound library consisting of DrugBank drugs including 1347 FDA approved, non-nutraceutical molecules. Twenty nine drugs ranked in the top 1 % for binding to each target were subsequently included in our experimental analysis. Continuous exposure of rotifers to 1 µM naproxen significantly extended rotifer mean lifespan by 14 %. We used three endpoints to estimate rotifer health: swimming speed (mobility proxy), reproduction (overall vitality), and mitochondria activity (cellular senescence proxy). The natural decline in swimming speed with aging was more gradual when rotifers were exposed to three drugs, so that on day 6, mean swimming speed of females was 1.19 mm/s for naproxen (P = 0.038), 1.20 for fludarabine (P = 0.040), 1.35 for hydralazine (P = 0.038), as compared to 0.88 mm/s in the control. The average reproduction of control females in the second half of their reproductive lifespan was 1.08 per day. In contrast, females treated with 1 µM naproxen produced 1.4 offspring per day (P = 0.027) and females treated with 10 µM fludarabine or 1 µM hydralazine produced 1.72 (P = <0.001) and 1.66 (P = 0.001) offspring per day, respectively. Mitochondrial activity naturally declines with rotifer aging, but B. manjavacas treated with 1 µM hydralazine or 10 µM fludarabine retained 49 % (P = 0.038) and 89 % (P = 0.002) greater mitochondria activity, respectively, than untreated controls. Our results demonstrate that coupling computation to experimentation can quickly identify new drug candidates with anti-aging potential. Screening drugs for anti-aging effects using a rotifer bioassay is a powerful first step in identifying compounds worthy of follow-up in vertebrate models. Even if lifespan extension is not observed, certain drugs could improve healthspan, slowing age-dependent losses in mobility and vitality.
Collapse
Affiliation(s)
- Terry W Snell
- School of Biology, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA.
| | - Rachel K Johnston
- School of Biology, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA
| | - Bharath Srinivasan
- School of Biology, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA
| | - Hongyi Zhou
- School of Biology, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA
| | - Mu Gao
- School of Biology, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA
| | - Jeffrey Skolnick
- School of Biology, Georgia Institute of Technology, Atlanta, GA, 30332-0230, USA
| |
Collapse
|
22
|
Metcalfe C, Ramasubramoni A, Pula G, Harper MT, Mundell SJ, Coxon CH. Thioredoxin Inhibitors Attenuate Platelet Function and Thrombus Formation. PLoS One 2016; 11:e0163006. [PMID: 27716777 PMCID: PMC5055343 DOI: 10.1371/journal.pone.0163006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 08/31/2016] [Indexed: 12/14/2022] Open
Abstract
Thioredoxin (Trx) is an oxidoreductase with important physiological function. Imbalances in the NADPH/thioredoxin reductase/thioredoxin system are associated with a number of pathologies, particularly cancer, and a number of clinical trials for thioredoxin and thioredoxin reductase inhibitors have been carried out or are underway. Due to the emerging role and importance of oxidoreductases for haemostasis and the current interest in developing inhibitors for clinical use, we thought it pertinent to assess whether inhibition of the NADPH/thioredoxin reductase/thioredoxin system affects platelet function and thrombosis. We used small molecule inhibitors of Trx (PMX 464 and PX-12) to determine whether Trx activity influences platelet function, as well as an unbiased proteomics approach to identify potential Trx substrates on the surface of platelets that might contribute to platelet reactivity and function. Using LC-MS/MS we found that PMX 464 and PX-12 affected the oxidation state of thiols in a number of cell surface proteins. Key surface receptors for platelet adhesion and activation were affected, including the collagen receptor GPVI and the von Willebrand factor receptor, GPIb. To experimentally validate these findings we assessed platelet function in the presence of PMX 464, PX-12, and rutin (a selective inhibitor of the related protein disulphide isomerase). In agreement with the proteomics data, small molecule inhibitors of thioredoxin selectively inhibited GPVI-mediated platelet activation, and attenuated ristocetin-induced GPIb-vWF-mediated platelet agglutination, thus validating the findings of the proteomics study. These data reveal a novel role for thioredoxin in regulating platelet reactivity via proteins required for early platelet responses at sites of vessel injury (GPVI and GPIb). This work also highlights a potential opportunity for repurposing of PMX 464 and PX-12 as antiplatelet agents.
Collapse
Affiliation(s)
- Clive Metcalfe
- Oxford Molecular and Pathology Institute, South Parks Road, Oxford, OX1 3RE, United Kingdom
| | - Anjana Ramasubramoni
- Oxford Molecular and Pathology Institute, South Parks Road, Oxford, OX1 3RE, United Kingdom
| | - Giordano Pula
- Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath, BA2 7AY, United Kingdom
| | - Matthew T. Harper
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, United Kingdom
| | - Stuart J. Mundell
- Department of Physiology, Pharmacology and Neuroscience, Medical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, United Kingdom
- * E-mail:
| | - Carmen H. Coxon
- Department of Physiology, Pharmacology and Neuroscience, Medical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, United Kingdom
| |
Collapse
|
23
|
Jiang P, Loyau S, Tchitchinadze M, Ropers J, Jondeau G, Jandrot-Perrus M. Inhibition of Glycoprotein VI Clustering by Collagen as a Mechanism of Inhibiting Collagen-Induced Platelet Responses: The Example of Losartan. PLoS One 2015; 10:e0128744. [PMID: 26052700 PMCID: PMC4460036 DOI: 10.1371/journal.pone.0128744] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 04/30/2015] [Indexed: 11/30/2022] Open
Abstract
Exposure of platelets to collagen triggers the formation of a platelet clot. Pharmacological agents capable of inhibiting platelet activation by collagen are thus of potential therapeutic interest. Thrombus formation is initiated by the interaction of the GPIb-V-IX complex with collagen-bound vWF, while GPVI interaction with collagen triggers platelet activation that is reinforced by ADP and thromboxane A2. Losartan is an angiotensin II (Ang II) type I receptor (AT1R) antagonist proposed to have an antiplatelet activity via the inhibition of both the thromboxane A2 (TXA2) receptor (TP) and the glycoprotein VI (GPVI). Here, we characterized in vitro the effects of losartan at different doses on platelet responses: losartan inhibited platelet aggregation and secretion induced by 1 μg.mL-1 and 10 μg.mL-1 of collagen with an IC50 of ~ 6 μM. Losartan inhibited platelet responses induced by the GPVI specific collagen related peptide but not by the α2β1 specific peptide. However, losartan did not inhibit the binding of recombinant GPVI to collagen, which is not in favor of a simple competition. Indeed, the clustering of GPVI observed in flow cytometry and using the Duolink methodology, was inhibited by losartan. The impact of a therapeutic dose of losartan (100 mg/day) on platelet responses was analyzed ex vivo in a double blind study. No statistically significant differences were observed between losartan-treated (n=25) and non-treated (n=30) patients in terms of collagen and U46619-induced platelet activation. These data indicate that in treated patients, losartan does not achieve a measurable antiplatelet effect but provide the proof of concept that inhibiting collagen-induced GPVI clustering is of pharmacological interest to obtain an antithrombotic efficacy.
Collapse
Affiliation(s)
- Peng Jiang
- Inserm, UMR_S1148, Paris, France
- Univ Paris Diderot, Sorbonne Paris Cité, UMR_S1148, Paris, France
| | | | - Maria Tchitchinadze
- APHP- CNMR Syndrome de Marfan et apparentés, Service de Cardiologie, Hôpital Bichat, Paris, France
| | - Jacques Ropers
- Unité de Recherche Clinique, Hôpital Ambroise Paré—UFR Médecine Paris-Ile-de-France-Ouest, Université Versailles St-Quentin, Boulogne, France
| | - Guillaume Jondeau
- Inserm, UMR_S1148, Paris, France
- Univ Paris Diderot, Sorbonne Paris Cité, UMR_S1148, Paris, France
- APHP- CNMR Syndrome de Marfan et apparentés, Service de Cardiologie, Hôpital Bichat, Paris, France
| | - Martine Jandrot-Perrus
- Inserm, UMR_S1148, Paris, France
- Univ Paris Diderot, Sorbonne Paris Cité, UMR_S1148, Paris, France
- * E-mail:
| |
Collapse
|
24
|
Abstract
The current standard care for acute coronary syndromes is dual antiplatelet therapy combining the COX1 inhibitor aspirin with a drug targeting the P2Y12 receptor, together with anticoagulation during and after early revascularization by percutaneous intervention. In very high-risk patients, glycoprotein (GP) IIb/IIIa antagonists may also be used. Secondary prevention of ischemic events requires dual antiplatelet therapy for several months followed by lifelong low-dose aspirin. The duration of treatment and the drugs to combine nevertheless remain matters of debate and the focus of ongoing research. Despite great progress, there is still room for improved efficacy and this could involve new targets for both antiplatelet drugs (like the thrombin receptor PAR1) and anticoagulants. However, improved efficacy is offset by an increased risk of bleeding. Stroke patients are still waiting for better treatment, their bleeding risk being particularly high. New targets including the collagen receptor, glycoprotein VI (GPVI), and the GPIb-von Willebrand factor axis, governing platelet interaction with the diseased vessel wall, should enable us to complete the armamentarium of antiplatelet drugs.
Collapse
Affiliation(s)
- C Gachet
- UMR_S949, INSERM, Strasbourg, France
- Etablissement Français du Sang-Alsace (EFS-Alsace), Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| |
Collapse
|