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; 45:4582-4597. [PMID: 39150906 PMCID: PMC11560278 DOI: 10.1093/eurheartj/ehae482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [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
|
Slater A, Khattak S, Thomas MR. GPVI inhibition: Advancing antithrombotic therapy in cardiovascular disease. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2024; 10:465-473. [PMID: 38453424 PMCID: PMC11323372 DOI: 10.1093/ehjcvp/pvae018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/08/2024] [Accepted: 03/05/2024] [Indexed: 03/09/2024]
Abstract
Glycoprotein (GP) VI (GPVI) plays a major role in thrombosis but not haemostasis, making it a promising antithrombotic target. The primary role of GPVI on the surface of platelets is a signalling receptor for collagen, which is one of the most potent thrombotic sub-endothelial components that is exposed by atherosclerotic plaque rupture. Inhibition of GPVI has therefore been investigated as a strategy for treatment and prevention of atherothrombosis, such as during stroke and acute coronary syndromes. A range of specific GPVI inhibitors have been characterized, and two of these inhibitors, glenzocimab and revacept, have completed Phase II clinical trials in ischaemic stroke. In this review, we summarize mechanisms of GPVI activation and the latest progress of clinically tested GPVI inhibitors, including their mechanisms of action. By focusing on what is known about GPVI activation, we also discuss whether alternate strategies could be used to target GPVI.
Collapse
Affiliation(s)
- Alexandre Slater
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, Birmingham, UK
| | - Sophia Khattak
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, Birmingham, UK
- Cardiology Department, Queen Elizabeth Hospital, University Hospitals Birmingham, B15 2GW, Birmingham, UK
| | - Mark R Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, Birmingham, UK
- Cardiology Department, Queen Elizabeth Hospital, University Hospitals Birmingham, B15 2GW, Birmingham, UK
| |
Collapse
|
3
|
Xu RG, Tiede C, Calabrese AN, Cheah LT, Adams TL, Gauer JS, Hindle MS, Webb BA, Yates DM, Slater A, Duval C, Naseem KM, Herr AB, Tomlinson DC, Watson SP, Ariëns RAS. Affimer reagents as tool molecules to modulate platelet GPVI-ligand interactions and specifically bind GPVI dimer. Blood Adv 2024; 8:3917-3928. [PMID: 38838227 PMCID: PMC11321386 DOI: 10.1182/bloodadvances.2024012689] [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] [Received: 01/17/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/07/2024] Open
Abstract
ABSTRACT Glycoprotein VI (GPVI) plays a key role in collagen-induced platelet aggregation. Affimers are engineered binding protein alternatives to antibodies. We screened and characterized GPVI-binding Affimers as novel tools to probe GPVI function. Among the positive clones, M17, D22, and D18 bound GPVI with the highest affinities (dissociation constant (KD) in the nanomolar range). These Affimers inhibited GPVI-collagen-related peptide (CRP)-XL/collagen interactions, CRP-XL/collagen-induced platelet aggregation, and D22 also inhibited in vitro thrombus formation on a collagen surface under flow. D18 bound GPVI dimer but not monomer. GPVI binding was increased for D18 but not M17/D22 upon platelet activation by CRP-XL and adenosine 5'-diphosphate. D22 but not M17/D18 displaced nanobody 2 (Nb2) binding to GPVI, indicating similar epitopes for D22 with Nb2 but not for M17/D18. Mapping of binding sites revealed that D22 binds a site that overlaps with Nb2 on the D1 domain, whereas M17 targets a site on the D2 domain, overlapping in part with the glenzocimab binding site, a humanized GPVI antibody fragment antigen-binding fragment. D18 targets a new region on the D2 domain. We found that D18 is a stable noncovalent dimer and forms a stable complex with dimeric GPVI with 1:1 stoichiometry. Taken together, our data demonstrate that Affimers modulate GPVI-ligand interactions and bind different sites on GPVI D1/D2 domains. D18 is dimer-specific and could be used as a tool to detect GPVI dimerization or clustering in platelets. A dimeric epitope regulating ligand binding was identified on the GPVI D2 domain, which could be used for the development of novel bivalent antithrombotic agents selectively targeting GPVI dimer on platelets.
Collapse
Affiliation(s)
- Rui-Gang Xu
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Christian Tiede
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Antonio N. Calabrese
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Lih T. Cheah
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Thomas L. Adams
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Julia S. Gauer
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Matthew S. Hindle
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
- Centre for Biomedical Science Research, School of Health, Leeds Beckett University, Leeds, United Kingdom
| | - Beth A. Webb
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Daisie M. Yates
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Alexandre Slater
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Cédric Duval
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Khalid M. Naseem
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Andrew B. Herr
- Division of Immunobiology and Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Darren C. Tomlinson
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Steve P. Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Robert A. S. Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
4
|
Wang X, Slater A, Lee SC, Harrison N, Pollock NL, Bakker SE, Navarro S, Nieswandt B, Dafforn TR, García Á, Watson SP, Tomlinson MG. Purification and characterisation of the platelet-activating GPVI/FcRγ complex in SMALPs. Arch Biochem Biophys 2024; 754:109944. [PMID: 38395124 DOI: 10.1016/j.abb.2024.109944] [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: 11/30/2023] [Revised: 02/13/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
The collagen/fibrin(ogen) receptor, glycoprotein VI (GPVI), is a platelet activating receptor and a promising anti-thrombotic drug target. However, while agonist-induced GPVI clustering on platelet membranes has been shown to be essential for its activation, it is unknown if GPVI dimerisation represents a unique conformation for ligand binding. Current GPVI structures all contain only the two immunoglobulin superfamily (IgSF) domains in the GPVI extracellular region, so lacking the mucin-like stalk, transmembrane, cytoplasmic tail of GPVI and its associated Fc receptor γ (FcRγ) homodimer signalling chain, and provide contradictory insights into the mechanisms of GPVI dimerisation. Here, we utilised styrene maleic-acid lipid particles (SMALPs) to extract GPVI in complex with its two associated FcRγ chains from transfected HEK-293T cells, together with the adjacent lipid bilayer, then purified and characterised the GPVI/FcRγ-containing SMALPs, to enable structural insights into the full-length GPVI/FcRγ complex. Using size exclusion chromatography followed by a native polyacrylamide gel electrophoresis (PAGE) method, SMA-PAGE, we revealed multiple sizes of the purified GPVI/FcRγ SMALPs, suggesting the potential existence of GPVI oligomers. Importantly, GPVI/FcRγ SMALPs were functional as they could bind collagen. Mono-dispersed GPVI/FcRγ SMALPs could be observed under negative stain electron microscopy. These results pave the way for the future investigation of GPVI stoichiometry and structure, while also validating SMALPs as a promising tool for the investigation of human membrane protein interactions, stoichiometry and structure.
Collapse
Affiliation(s)
- Xueqing Wang
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, and Instituto de Investigación Sanitaria de Santiago (IDIS), 15782 Santiago de Compostela, Spain.
| | - Alexandre Slater
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), The Universities of Birmingham and Nottingham, The Midlands, UK
| | - Sarah C Lee
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Neale Harrison
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Naomi L Pollock
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Saskia E Bakker
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Stefano Navarro
- Institute of Experimental Biomedicine I, University Hospital Würzburg, Würzburg Josef-Schneider-Straße 2, 97080 Wurzburg, Germany; Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, 97080 Wurzburg, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine I, University Hospital Würzburg, Würzburg Josef-Schneider-Straße 2, 97080 Wurzburg, Germany; Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, 97080 Wurzburg, Germany
| | - Tim R Dafforn
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Ángel García
- Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, and Instituto de Investigación Sanitaria de Santiago (IDIS), 15782 Santiago de Compostela, Spain
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), The Universities of Birmingham and Nottingham, The Midlands, UK
| | - Michael G Tomlinson
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), The Universities of Birmingham and Nottingham, The Midlands, UK.
| |
Collapse
|
5
|
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
|
6
|
Navarro S, Vögtle T, Groß N, Preu J, Englert M, Nieswandt B, Bösl MR, Stegner D. Mutations of the dimerization site of glycoprotein (GP) VI result in abolished expression. Thromb Res 2023; 232:89-92. [PMID: 37951045 DOI: 10.1016/j.thromres.2023.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/13/2023]
Affiliation(s)
- Stefano Navarro
- Julius-Maximilians-Universität Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany; University Hospital Würzburg, Institute of Experimental Biomedicine, Würzburg, Germany
| | - Timo Vögtle
- Julius-Maximilians-Universität Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany; University Hospital Würzburg, Institute of Experimental Biomedicine, Würzburg, Germany
| | - Nina Groß
- University Hospital Würzburg, Institute of Experimental Biomedicine, Würzburg, Germany
| | - Julia Preu
- Julius-Maximilians-Universität Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany; University Hospital Würzburg, Institute of Experimental Biomedicine, Würzburg, Germany
| | - Maximilian Englert
- University Hospital Würzburg, Institute of Experimental Biomedicine, Würzburg, Germany
| | - Bernhard Nieswandt
- Julius-Maximilians-Universität Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany; University Hospital Würzburg, Institute of Experimental Biomedicine, Würzburg, Germany
| | - Michael R Bösl
- Julius-Maximilians-Universität Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany; University Hospital Würzburg, Institute of Experimental Biomedicine, Würzburg, Germany.
| | - David Stegner
- Julius-Maximilians-Universität Würzburg, Rudolf Virchow Center for Integrative and Translational Bioimaging, Würzburg, Germany; University Hospital Würzburg, Institute of Experimental Biomedicine, Würzburg, Germany.
| |
Collapse
|
7
|
Yasmin R, Chanchal S, Ashraf MZ, Doley R. Daboxin P, a phospholipase A 2 of Indian Daboia russelii venom, modulates thrombin-mediated platelet aggregation. J Biochem Mol Toxicol 2023; 37:e23476. [PMID: 37466159 DOI: 10.1002/jbt.23476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/12/2023] [Accepted: 07/08/2023] [Indexed: 07/20/2023]
Abstract
Daboxin P, reported earlier from the venom of Daboia russellii, disturbs the blood coagulation cascade by targeting factor X and factor Xa. The present study exhibits that Daboxin P also inhibits platelet aggregation induced by various agonists. The thrombin-induced platelet aggregation was inhibited maximum whereas inhibition of collagen-induced platelet aggregation was found to be 50% and no inhibition of adenosine diphosphate (ADP) and arachidonic acid-induced aggregation was observed. Daboxin P dose-dependently inhibited the thrombin-induced platelet aggregation with Anti-Aggregation 50 (AD50 ) dose of 55.166 nM and also reduced the thrombin-mediated calcium influx. In-silico interaction studies suggested that Daboxin P binds to thrombin and blocks its interaction with its receptor on the platelet surface. Quenching of thrombin's emission spectrum by Daboxin P and electrophoretic profiles of pull-down assay further reveals the binding between Daboxin P and thrombin. Thus, the present study demonstrates that Daboxin P inhibits thrombin-induced platelet aggregation by binding to thrombin.
Collapse
Affiliation(s)
- Rafika Yasmin
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, India
| | - Shankar Chanchal
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi, New Delhi, India
| | - Mohammad Zahid Ashraf
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi, New Delhi, India
| | - Robin Doley
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, India
| |
Collapse
|
8
|
Ye Y, Yang L, Leng M, Wang Q, Wu J, Wan W, Wang H, Li L, Peng Y, Chai S, Meng Z. Luteolin inhibits GPVI-mediated platelet activation, oxidative stress, and thrombosis. Front Pharmacol 2023; 14:1255069. [PMID: 38026984 PMCID: PMC10644720 DOI: 10.3389/fphar.2023.1255069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction: Luteolin inhibits platelet activation and thrombus formation, but the mechanisms are unclear. This study investigated the effects of luteolin on GPVI-mediated platelet activation in vitro and explored the effect of luteolin on thrombosis, coagulation, and platelet production in vivo. Methods: Washed human platelets were used for aggregation, membrane protein expression, ATP, Ca2+, and LDH release, platelet adhesion/spreading, and clot retraction experiments. Washed human platelets were used to detect collagen and convulxin-induced reactive oxygen species production and endogenous antioxidant effects. C57BL/6 male mice were used for ferric chloride-induced mesenteric thrombosis, collagen-epinephrine induced acute pulmonary embolism, tail bleeding, coagulation function, and luteolin toxicity experiments. The interaction between luteolin and GPVI was analyzed using solid phase binding assay and surface plasmon resonance (SPR). Results: Luteolin inhibited collagen- and convulxin-mediated platelet aggregation, adhesion, and release. Luteolin inhibited collagen- and convulxin-induced platelet ROS production and increased platelet endogenous antioxidant capacity. Luteolin reduced convulxin-induced activation of ITAM and MAPK signaling molecules. Molecular docking simulation showed that luteolin forms hydrogen bonds with GPVI. The solid phase binding assay showed that luteolin inhibited the interaction between collagen and GPVI. Surface plasmon resonance showed that luteolin bonded GPVI. Luteolin inhibited integrin αIIbβ3-mediated platelet activation. Luteolin inhibited mesenteric artery thrombosis and collagen- adrenergic-induced pulmonary thrombosis in mice. Luteolin decreased oxidative stress in vivo. Luteolin did not affect coagulation, hemostasis, or platelet production in mice. Discussion: Luteolin may be an effective and safe antiplatelet agent target for GPVI. A new mechanism (decreased oxidative stress) for the anti-platelet activity of luteolin has been identified.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Zhaohui Meng
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
9
|
Hou C, Lei Y, Li N, Wei M, Wang S, Rahman SU, Bao C, Bao B, Elango J, Wu W. Collagen from Iris squid grafted with polyethylene glycol and collagen peptides promote the proliferation of fibroblast through PI3K/AKT and Ras/RAF/MAPK signaling pathways. Int J Biol Macromol 2023; 247:125772. [PMID: 37429348 DOI: 10.1016/j.ijbiomac.2023.125772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/02/2023] [Accepted: 07/07/2023] [Indexed: 07/12/2023]
Abstract
Collagens from marine sources have been used widely in food, cosmetics and tissue engineering application due to their excellent functional and biological properties. In the present study, a novel protein, collagen from iris squid skin (SSC) was characterized, grafted with polyethylene-glycol (PEG) and Acid-Green 20 (AG) and was investigated the molecular signaling pathways in L-929 fibroblast cells along with their structural peptide analogs. SDS-PAGE and IR spectrum of SSC analysis showed the typical structure of type I collagen. The fibroblast proliferation was evaluated for SSC, SSC grafted PEG (SSC-PEG) and their structural analogs including Gly-Pro-Leu-Gly-Leu-Leu (PEP1), Gly-Pro-Leu-Gly-Leu-Leu-Gly-Phe-Leu (PEP2), Gly-Pro-Leu-Gly-Leu-Leu-Gly-Phe-Leu-Gly-Pro-Leu (PEP3) and Gly-Pro-Leu-Gly-Leu-Leu-Gly-Phe-Leu-Gly-Pro-Leu-Gly-Leu-Ser (PEP4). The optimal concentration of SSC and its derivative was 0.07 μ mol/L. The fibroblast growth-promoting factors were promoted by all the treatment groups by accelerating the PI3K/AKT and Ras/RAF/MAPK signaling pathways in L-929 cells, and inhibiting the secretion of apoptotic factors. Compared to the control group, mRNA and protein expression of AKT in the PI3K/AKT and Ras in Ras/RAF/MAPK signaling pathway were accelerated significantly by PEP4, respectively, while the Bax value was significantly lower (P < 0.01). The promoting effect of PEP1, PEP2, PEP3 and PEP4 on L-929 cells was closely related to the length of the peptides. Therefore, this study disclosed that PEP1, PEP2, PEP3 and PEP4 were novel analogs that greatly promote the proliferation of L-929 cells through PI3K/AKT and Ras/RAF/MAPK signaling pathways.
Collapse
Affiliation(s)
- Chunyu Hou
- Department of Marine Bio-pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Yunjia Lei
- Department of Marine Bio-pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China.
| | - Na Li
- Department of Marine Bio-pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Mingjun Wei
- Department of Marine Bio-pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China.
| | - Shujun Wang
- Department of Marine Biopharmacology, College of Food Science and Technology, Jiangsu Ocean University, Lianyungang City 222005, Jiangsu Province, China
| | - Saeed Ur Rahman
- Oral Biology, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar 25000, Pakistan
| | - Chunling Bao
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 201306, China.
| | - Bin Bao
- Zhoushan Marine Biological Engineering Co., Ltd, Zhoushan City 316104, Zhejiang Province, China.
| | - Jeevithan Elango
- Department of Marine Bio-pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China; Center of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India; Department of Biomaterials Engineering, Faculty of Health Sciences, UCAM- Universidad Católica San Antonio de Murcia, Guadalupe, 30107, Murcia, Spain.
| | - Wenhui Wu
- Department of Marine Bio-pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
10
|
AlOuda SK, Sasikumar P, AlThunayan T, Alaajam F, Khan S, Sahli KA, Abohassan MS, Pollitt A, Jung SM, Gibbins JM. Role of heat shock protein 47 in platelet glycoprotein VI dimerization and signaling. Res Pract Thromb Haemost 2023; 7:102177. [PMID: 37767064 PMCID: PMC10520510 DOI: 10.1016/j.rpth.2023.102177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/23/2022] [Accepted: 07/21/2023] [Indexed: 09/29/2023] Open
Abstract
Background Heat shock protein 47 (HSP47) is an intracellular chaperone protein with an indispensable role in collagen biosynthesis in collagen-secreting cells. This chaperone has also been shown to be released and present on the surface of platelets. The inhibition of HSP47 in human platelets or its ablation in mouse platelets reduces platelet function in response to collagen and the glycoprotein (GP) VI collagen receptor agonist CRP-XL. Objectives In this study, we sought, through experiments, to explore cellular distribution, trafficking, and influence on GPVI interactions to understand how HSP47 modulates collagen receptor signaling. Methods HSP47-deficient mouse platelets and SMIH- treated human platelets were used to study the role of HSP47 in collagen mediated responses and signaling. Results Using subcellular fractionation analysis and immunofluorescence microscopy, HSP47 was found to be localized to the platelet-dense tubular system. Following platelet stimulation, HSP47 mobilization to the cell surface was shown to be dependent on actin polymerization, a feature common to other dense tubular system resident platelet proteins that are released to the cell surface during activation. In this location, HSP47 was found to contribute to platelet adhesion to collagen or CRP-XL but not to GFOGER peptide (an integrin α2β1-binding sequence within collagens), indicating selective effects of HSP47 on GPVI function. Dimerization of GPVI on the platelet surface increases its affinity for collagen. GPVI dimerization was reduced following HSP47 inhibition, as was collagen and CRP-XL-mediated signaling. Conclusion The present study identifies a role for cell surface-localized HSP47 in modulating platelet responses to collagen through dimerization of GPVI, thereby enhancing platelet signaling and activation.
Collapse
Affiliation(s)
- Sarah K. AlOuda
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Parvathy Sasikumar
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
- Centre for Haematology, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Taysseer AlThunayan
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Fahd Alaajam
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Gizan, Saudi Arabia
| | - Sabeeya Khan
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Khaled A. Sahli
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
- General Directorate of Medical Services, Ministry of Interior, Riyadh, Kingdom of Saudi Arabia
| | - Mohammed S. Abohassan
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Alice Pollitt
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Stephanie M. Jung
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Jonathan M. Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| |
Collapse
|
11
|
Mangin PH, Gardiner EE, Ariëns RAS, Jandrot-Perrus M. Glycoprotein VI interplay with fibrin(ogen) in thrombosis. J Thromb Haemost 2023; 21:1703-1713. [PMID: 36990158 DOI: 10.1016/j.jtha.2023.03.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Platelets play a central role in the arrest of bleeding. The ability of platelets to engage with extracellular matrix proteins of the subendothelium has long been recognized as a pivotal platelet attribute, underpinning adequate hemostasis. The propensity of platelets to rapidly bind and functionally respond to collagen was one of the earliest documented events in platelet biology. The receptor primarily responsible for mediating platelet/collagen responses was identified as glycoprotein (GP) VI and successfully cloned in 1999. Since that time, this receptor has held the attention of many research groups, and through these efforts, we now have an excellent understanding of the roles of GPVI as a platelet- and megakaryocyte-specific adheso-signaling receptor in platelet biology. GPVI is considered a viable antithrombotic target, as data obtained from groups across the world is consistent with GPVI being less involved in physiological hemostatic processes but participating in arterial thrombosis. This review will highlight the key aspects of GPVI contributions to platelet biology and concentrate on the interaction with recently identified ligands, with a focus on fibrin and fibrinogen, discussing the role of these interactions in the growth and stability of thrombi. We will also discuss important therapeutic developments that target GPVI to modulate platelet function while minimizing bleeding outcomes.
Collapse
Affiliation(s)
- Pierre H Mangin
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale, Etablissement Français du Sang Grand-Est, Unité Mixte de Recherche-S1255, Fédération de Médecine Translationnelle de Strasbourg F-67065 Strasbourg, France.
| | - Elizabeth E Gardiner
- The John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Robert A S Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Martine Jandrot-Perrus
- Université de Paris Institut National de la Santé et de la Recherche Médicale, UMR-S1148, Hôpital Bichat, Paris, France
| |
Collapse
|
12
|
Billiald P, Slater A, Welin M, Clark JC, Loyau S, Pugnière M, Jiacomini IG, Rose N, Lebozec K, Toledano E, François D, Watson SP, Jandrot-Perrus M. Targeting platelet GPVI with glenzocimab: a novel mechanism for inhibition. Blood Adv 2023; 7:1258-1268. [PMID: 36375047 PMCID: PMC10119634 DOI: 10.1182/bloodadvances.2022007863] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
Platelet glycoprotein VI (GPVI) is attracting interest as a potential target for the development of new antiplatelet molecules with a low bleeding risk. GPVI binding to vascular collagen initiates thrombus formation and GPVI interactions with fibrin promote the growth and stability of the thrombus. In this study, we show that glenzocimab, a clinical stage humanized antibody fragment (Fab) with a high affinity for GPVI, blocks the binding of both ligands through a combination of steric hindrance and structural change. A cocrystal of glenzocimab with an extracellular domain of monomeric GPVI was obtained and its structure determined to a resolution of 1.9 Å. The data revealed that (1) glenzocimab binds to the D2 domain of GPVI, GPVI dimerization was not observed in the crystal structure because glenzocimab prevented D2 homotypic interactions and the formation of dimers that have a high affinity for collagen and fibrin; and (2) the light variable domain of the GPVI-bound Fab causes steric hindrance that is predicted to prevent the collagen-related peptide (CRP)/collagen fibers from extending out of their binding site and preclude GPVI clustering and downstream signaling. Glenzocimab did not bind to a truncated GPVI missing loop residues 129 to 136, thus validating the epitope identified in the crystal structure. Overall, these findings demonstrate that the binding of glenzocimab to the D2 domain of GPVI induces steric hindrance and structural modifications that drive the inhibition of GPVI interactions with its major ligands.
Collapse
Affiliation(s)
- Philippe Billiald
- Laboratory for Vascular Translational Science, UMR_S1148 INSERM, Université Paris Cité, Hôpital Bichat, Paris, France
- School of Pharmacy, Université Paris-Saclay, Orsay, France
| | - Alexandre Slater
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Martin Welin
- SARomics Biostructures, Medicon Village, Lund, Sweden
| | - Joanne C. Clark
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Stéphane Loyau
- Laboratory for Vascular Translational Science, UMR_S1148 INSERM, Université Paris Cité, Hôpital Bichat, Paris, France
| | - Martine Pugnière
- Institut de Recherche en Cancérologie de Montpellier, INSERM, U1194, Université Montpellier, ICM Institut Régional du Cancer, Montpellier, France
| | - Isabella G. Jiacomini
- Departamento de Patologia Básica, Laboratório de Imunoquímica, Universidade Federal do Paraná, Curitiba, Brazil
| | - Nadia Rose
- SARomics Biostructures, Medicon Village, Lund, Sweden
| | | | | | | | - Steve P. Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Midlands, UK
| | - Martine Jandrot-Perrus
- Laboratory for Vascular Translational Science, UMR_S1148 INSERM, Université Paris Cité, Hôpital Bichat, Paris, France
| |
Collapse
|
13
|
Tantiwong C, Dunster JL, Cavill R, Tomlinson MG, Wierling C, Heemskerk JWM, Gibbins JM. An agent-based approach for modelling and simulation of glycoprotein VI receptor diffusion, localisation and dimerisation in platelet lipid rafts. Sci Rep 2023; 13:3906. [PMID: 36890261 PMCID: PMC9994409 DOI: 10.1038/s41598-023-30884-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 03/02/2023] [Indexed: 03/10/2023] Open
Abstract
Receptor diffusion plays an essential role in cellular signalling via the plasma membrane microenvironment and receptor interactions, but the regulation is not well understood. To aid in understanding of the key determinants of receptor diffusion and signalling, we developed agent-based models (ABMs) to explore the extent of dimerisation of the platelet- and megakaryocyte-specific receptor for collagen glycoprotein VI (GPVI). This approach assessed the importance of glycolipid enriched raft-like domains within the plasma membrane that lower receptor diffusivity. Our model simulations demonstrated that GPVI dimers preferentially concentrate in confined domains and, if diffusivity within domains is decreased relative to outside of domains, dimerisation rates are increased. While an increased amount of confined domains resulted in further dimerisation, merging of domains, which may occur upon membrane rearrangements, was without effect. Modelling of the proportion of the cell membrane which constitutes lipid rafts indicated that dimerisation levels could not be explained by these alone. Crowding of receptors by other membrane proteins was also an important determinant of GPVI dimerisation. Together, these results demonstrate the value of ABM approaches in exploring the interactions on a cell surface, guiding the experimentation for new therapeutic avenues.
Collapse
Affiliation(s)
- Chukiat Tantiwong
- School of Biological Sciences, University of Reading, Reading, UK.,Department of Biochemistry, CARIM, Maastricht University, Maastricht, The Netherlands
| | - Joanne L Dunster
- School of Biological Sciences, University of Reading, Reading, UK
| | - Rachel Cavill
- Department of Data Science and Knowledge Engineering, Maastricht University, Maastricht, The Netherlands
| | | | | | - Johan W M Heemskerk
- Department of Biochemistry, CARIM, Maastricht University, Maastricht, The Netherlands.,Synapse Research Institute, Maastricht, The Netherlands
| | | |
Collapse
|
14
|
Han C, Ren P, Mamtimin M, Kruk L, Sarukhanyan E, Li C, Anders HJ, Dandekar T, Krueger I, Elvers M, Goebel S, Adler K, Münch G, Gudermann T, Braun A, Mammadova-Bach E. Minimal Collagen-Binding Epitope of Glycoprotein VI in Human and Mouse Platelets. Biomedicines 2023; 11:biomedicines11020423. [PMID: 36830959 PMCID: PMC9952969 DOI: 10.3390/biomedicines11020423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/22/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
Glycoprotein VI (GPVI) is a platelet-specific receptor for collagen and fibrin, regulating important platelet functions such as platelet adhesion and thrombus growth. Although the blockade of GPVI function is widely recognized as a potent anti-thrombotic approach, there are limited studies focused on site-specific targeting of GPVI. Using computational modeling and bioinformatics, we analyzed collagen- and CRP-binding surfaces of GPVI monomers and dimers, and compared the interacting surfaces with other mammalian GPVI isoforms. We could predict a minimal collagen-binding epitope of GPVI dimer and designed an EA-20 antibody that recognizes a linear epitope of this surface. Using platelets and whole blood samples donated from wild-type and humanized GPVI transgenic mice and also humans, our experimental results show that the EA-20 antibody inhibits platelet adhesion and aggregation in response to collagen and CRP, but not to fibrin. The EA-20 antibody also prevents thrombus formation in whole blood, on the collagen-coated surface, in arterial flow conditions. We also show that EA-20 does not influence GPVI clustering or receptor shedding. Therefore, we propose that blockade of this minimal collagen-binding epitope of GPVI with the EA-20 antibody could represent a new anti-thrombotic approach by inhibiting specific interactions between GPVI and the collagen matrix.
Collapse
Affiliation(s)
- Chao Han
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilian-University, 80336 Munich, Germany
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig-Maximilian-University, 80336 Munich, Germany
| | - Pengxuan Ren
- School of Life Science and Technology, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Medina Mamtimin
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilian-University, 80336 Munich, Germany
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig-Maximilian-University, 80336 Munich, Germany
| | - Linus Kruk
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilian-University, 80336 Munich, Germany
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig-Maximilian-University, 80336 Munich, Germany
| | - Edita Sarukhanyan
- Department of Bioinformatics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Chenyu Li
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig-Maximilian-University, 80336 Munich, Germany
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig-Maximilian-University, 80336 Munich, Germany
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Irena Krueger
- Department of Vascular and Endovascular Surgery, Heinrich-Heine University Medical Center, 40225 Düsseldorf, Germany
| | - Margitta Elvers
- Department of Vascular and Endovascular Surgery, Heinrich-Heine University Medical Center, 40225 Düsseldorf, Germany
| | | | | | - Götz Münch
- AdvanceCOR GmbH, 82152 Martinsried, Germany
| | - Thomas Gudermann
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilian-University, 80336 Munich, Germany
- German Center for Lung Research (DZL), 80336 Munich, Germany
| | - Attila Braun
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilian-University, 80336 Munich, Germany
- Correspondence: (A.B.); (E.M.-B.)
| | - Elmina Mammadova-Bach
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilian-University, 80336 Munich, Germany
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig-Maximilian-University, 80336 Munich, Germany
- Correspondence: (A.B.); (E.M.-B.)
| |
Collapse
|
15
|
Damaskinaki FN, Jooss NJ, Martin EM, Clark JC, Thomas MR, Poulter NS, Emsley J, Kellam B, Watson SP, Slater A. Characterizing the binding of glycoprotein VI with nanobody 35 reveals a novel monomeric structure of glycoprotein VI where the conformation of D1+D2 is independent of dimerization. J Thromb Haemost 2023; 21:317-328. [PMID: 36700508 DOI: 10.1016/j.jtha.2022.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/10/2022] [Accepted: 11/06/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND The platelet-signaling receptor glycoprotein VI (GPVI) is a promising antithrombotic target. We have previously raised a series of high-affinity nanobodies (Nbs) against GPVI and identified Nb2, Nb21, and Nb35 as potent GPVI inhibitors. The Nb2 binding site has been mapped to the D1 domain, which is directly adjacent to the CRP binding site. Ligand-binding complementary determining region 3 has only 15% conservation between all 3 Nbs. OBJECTIVES To map the binding sites of Nb21 and Nb35 on GPVI. METHODS We determined the X-ray crystal structure of the D1 and D2 extracellular domains of the GPVI-Nb35 complex. We then looked at the effects of various GPVI mutations on the ability of Nbs to inhibit collagen binding and GPVI signaling using surface binding assays and transfected cell lines. RESULTS The crystal structure of GPVI bound to Nb35 was solved. GPVI was present as a monomer, and the D1+D2 conformation was comparable to that in the dimeric structure. Arg46, Tyr47, and Ala57 are common residues on GPVI targeted by both Nb2 and Nb35. Mutating Arg46 to an Ala abrogated the ability of Nb2, Nb21, and Nb35 to inhibit collagen-induced GPVI signaling and blocked the binding of all 3 Nbs. In addition, Arg60 was found to reduce Nb21 inhibition but not the inhibition Nb2 or Nb35. CONCLUSIONS These findings reveal key residues involved in the high-affinity binding of GPVI inhibitors and negate the idea that GPVI dimerization induces a conformational change required for ligand binding.
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), Universities of Birmingham and Nottingham, The Midlands, UK; Biodiscovery Institute, University Park, University of Nottingham, Nottingham, UK
| | - Natalie J Jooss
- Institute of Cardiovascular Sciences, Level 1 IBR, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Eleyna M Martin
- Institute of Cardiovascular Sciences, Level 1 IBR, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Joanne C Clark
- 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), Universities of Birmingham and Nottingham, The Midlands, UK
| | - Mark R Thomas
- Institute of Cardiovascular Sciences, Level 1 IBR, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK; Department of Cardiology, University Hospitals Birmingham, Birmingham, UK
| | - Natalie S Poulter
- 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), Universities of Birmingham and Nottingham, The Midlands, UK
| | - Jonas Emsley
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, UK; Biodiscovery Institute, University Park, University of Nottingham, Nottingham, UK
| | - Barrie Kellam
- Centre of Membrane Proteins and Receptors (COMPARE), 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), Universities of Birmingham and Nottingham, The Midlands, UK
| | - Alexandre Slater
- Institute of Cardiovascular Sciences, Level 1 IBR, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
16
|
Artesunate as a glycoprotein VI antagonist for preventing platelet activation and thrombus formation. Biomed Pharmacother 2022; 153:113531. [DOI: 10.1016/j.biopha.2022.113531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 11/22/2022] Open
|
17
|
Modulation of Glycoprotein VI and Its Downstream Signaling Pathways as an Antiplatelet Target. Int J Mol Sci 2022; 23:ijms23179882. [PMID: 36077280 PMCID: PMC9456422 DOI: 10.3390/ijms23179882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 11/16/2022] Open
Abstract
Antiplatelet therapy aims to reduce the risk of thrombotic events while maintaining hemostasis. A promising current approach is the inhibition of platelet glycoprotein GPVI-mediated adhesion pathways; pathways that do not involve coagulation. GPVI is a signaling receptor integral for collagen-induced platelet activation and participates in the thrombus consolidation process, being a suitable target for thrombosis prevention. Considering this, the blocking or antibody-mediated depletion of GPVI is a promising antiplatelet therapy for the effective and safe treatment of thrombotic diseases without a significant risk of bleeding and impaired hemostatic plug formation. This review describes the current knowledge concerning pharmaceutical approaches to platelet GPVI modulation and its downstream signaling pathways in this context.
Collapse
|
18
|
GPVI and collagen: the final word? Blood 2022; 139:3005-3007. [PMID: 35587869 DOI: 10.1182/blood.2022015962] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 02/26/2022] [Indexed: 11/20/2022] Open
|
19
|
Structural insights into collagen binding by platelet receptor glycoprotein VI. Blood 2022; 139:3087-3098. [PMID: 35245360 DOI: 10.1182/blood.2021013614] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 02/08/2022] [Indexed: 02/02/2023] Open
Abstract
Glycoprotein VI (GPVI) mediates collagen-induced platelet activation after vascular damage and is an important contributor to the onset of thrombosis, heart attack, and stroke. Animal models of thrombosis have identified GPVI as a promising target for antithrombotic therapy. Although for many years the crystal structure of GPVI has been known, the essential details of its interaction with collagen have remained elusive. Here, we present crystal structures of the GPVI ectodomain bound to triple-helical collagen peptides, which reveal a collagen-binding site across the β-sheet of the D1 domain. Mutagenesis and binding studies confirm the observed binding site and identify Trp76, Arg38, and Glu40 as essential residues for binding to fibrillar collagens and collagen-related peptides (CRPs). GPVI binds a site on collagen comprising two collagen chains with the core formed by the sequence motif OGPOGP. Potent GPVI-binding peptides from Toolkit-III all contain OGPOGP; weaker binding peptides frequently contain a partial motif varying at either terminus. Alanine-scanning of peptide III-30 also identified two AGPOGP motifs that contribute to GPVI binding, but steric hindrance between GPVI molecules restricts the maximum binding capacity. We further show that no cooperative interactions could occur between two GPVI monomers binding to a stretch of (GPO)5 and that binding of ≥2 GPVI molecules to a fibril-embedded helix requires non-overlapping OGPOGP motifs. Our structure confirms the previously suggested similarity in collagen binding between GPVI and leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1) but also indicates significant differences that may be exploited for the development of receptor-specific therapeutics.
Collapse
|
20
|
Picker J, Lan Z, Arora S, Green M, Hahn M, Cosgriff-Hernandez E, Hook M. Prokaryotic Collagen-Like Proteins as Novel Biomaterials. Front Bioeng Biotechnol 2022; 10:840939. [PMID: 35372322 PMCID: PMC8968730 DOI: 10.3389/fbioe.2022.840939] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/10/2022] [Indexed: 12/13/2022] Open
Abstract
Collagens are the major structural component in animal extracellular matrices and are critical signaling molecules in various cell-matrix interactions. Its unique triple helical structure is enabled by tripeptide Gly-X-Y repeats. Understanding of sequence requirements for animal-derived collagen led to the discovery of prokaryotic collagen-like protein in the early 2000s. These prokaryotic collagen-like proteins are structurally similar to mammalian collagens in many ways. However, unlike the challenges associated with recombinant expression of mammalian collagens, these prokaryotic collagen-like proteins can be readily expressed in E. coli and are amenable to genetic modification. In this review article, we will first discuss the properties of mammalian collagen and provide a comparative analysis of mammalian collagen and prokaryotic collagen-like proteins. We will then review the use of prokaryotic collagen-like proteins to both study the biology of conventional collagen and develop a new biomaterial platform. Finally, we will describe the application of Scl2 protein, a streptococcal collagen-like protein, in thromboresistant coating for cardiovascular devices, scaffolds for bone regeneration, chronic wound dressing and matrices for cartilage regeneration.
Collapse
Affiliation(s)
- Jonathan Picker
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M, Houston, TX, United States
| | - Ziyang Lan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Srishtee Arora
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M, Houston, TX, United States
| | - Mykel Green
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Mariah Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| | | | - Magnus Hook
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M, Houston, TX, United States
| |
Collapse
|
21
|
Induruwa I, McKinney H, Kempster C, Thomas P, Batista J, Malcor JD, Bonna A, McGee J, Bumanlag-Amis E, Rehnstrom K, Ashford S, Soejima K, Ouwehand W, Farndale R, Downes K, Warburton E, Moroi M, Jung S. Platelet surface receptor glycoprotein VI-dimer is overexpressed in stroke: The Glycoprotein VI in Stroke (GYPSIE) study results. PLoS One 2022; 17:e0262695. [PMID: 35041713 PMCID: PMC8765640 DOI: 10.1371/journal.pone.0262695] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 01/03/2022] [Indexed: 11/30/2022] Open
Abstract
Objectives Platelet activation underpins thrombus formation in ischemic stroke. The active, dimeric form of platelet receptor glycoprotein (GP) VI plays key roles by binding platelet ligands collagen and fibrin, leading to platelet activation. We investigated whether patients presenting with stroke expressed more GPVI on their platelet surface and had more active circulating platelets as measured by platelet P-selectin exposure. Methods 129 ischemic or hemorrhagic stroke patients were recruited within 8h of symptom onset. Whole blood was analyzed for platelet-surface expression of total GPVI, GPVI-dimer, and P-selectin by flow cytometry at admission and day-90 post-stroke. Results were compared against a healthy control population (n = 301). Results The platelets of stroke patients expressed significantly higher total GPVI and GPVI-dimer (P<0.0001) as well as demonstrating higher resting P-selectin exposure (P<0.0001), a measure of platelet activity, compared to the control group, suggesting increased circulating platelet activation. GPVI-dimer expression was strongly correlated circulating platelet activation [r2 = 0.88, P<0.0001] in stroke patients. Furthermore, higher platelet surface GPVI expression was associated with increased stroke severity at admission. At day-90 post-stroke, GPVI-dimer expression and was further raised compared to the level at admission (P<0.0001) despite anti-thrombotic therapy. All ischemic stroke subtypes and hemorrhagic strokes expressed significantly higher GPVI-dimer compared to controls (P<0.0001). Conclusions Stroke patients express more GPVI-dimer on their platelet surface at presentation, lasting at least until day-90 post-stroke. Small molecule GPVI-dimer inhibitors are currently in development and the results of this study validate that GPVI-dimer as an anti-thrombotic target in ischemic stroke.
Collapse
Affiliation(s)
- Isuru Induruwa
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| | - Harriet McKinney
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Carly Kempster
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Patrick Thomas
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Joana Batista
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Jean-Daniel Malcor
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Arkadiusz Bonna
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Joanne McGee
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Elaine Bumanlag-Amis
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Karola Rehnstrom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Sophie Ashford
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Kenji Soejima
- Research and Development Coordination and Administration Department, KM Biologics Co., Ltd, Kumamoto, Japan
| | - Willem Ouwehand
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Richard Farndale
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Kate Downes
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Elizabeth Warburton
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Masaaki Moroi
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Stephanie Jung
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
22
|
Diindolylmethane ameliorates platelet aggregation and thrombosis: In silico, in vitro, and in vivo studies. Eur J Pharmacol 2022; 919:174812. [DOI: 10.1016/j.ejphar.2022.174812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/21/2022] [Accepted: 02/08/2022] [Indexed: 01/01/2023]
|
23
|
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: 0.7] [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]
|
24
|
Montague SJ, Patel P, Martin EM, Slater A, Quintanilla LG, Perrella G, Kardeby C, Nagy M, Mezzano D, Mendes PM, Watson SP. Platelet activation by charged ligands and nanoparticles: platelet glycoprotein receptors as pattern recognition receptors. Platelets 2021; 32:1018-1030. [PMID: 34266346 DOI: 10.1080/09537104.2021.1945571] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/10/2021] [Accepted: 06/15/2021] [Indexed: 02/08/2023]
Abstract
Charge interactions play a critical role in the activation of the innate immune system by damage- and pathogen-associated molecular pattern receptors. The ability of these receptors to recognize a wide spectrum of ligands through a common mechanism is critical in host defense. In this article, we argue that platelet glycoprotein receptors that signal through conserved tyrosine-based motifs function as pattern recognition receptors (PRRs) for charged endogenous and exogenous ligands, including sulfated polysaccharides, charged proteins and nanoparticles. This is exemplified by GPVI, CLEC-2 and PEAR1 which are activated by a wide spectrum of endogenous and exogenous ligands, including diesel exhaust particles, sulfated polysaccharides and charged surfaces. We propose that this mechanism has evolved to drive rapid activation of platelets at sites of injury, but that under some conditions it can drive occlusive thrombosis, for example, when blood comes into contact with infectious agents or toxins. In this Opinion Article, we discuss mechanisms behind charge-mediated platelet activation and opportunities for designing nanoparticles and related agents such as dendrimers as novel antithrombotics.
Collapse
Affiliation(s)
- Samantha J Montague
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Pushpa Patel
- School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, UK
| | - Eleyna M Martin
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Alexandre Slater
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Lourdes Garcia Quintanilla
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Gina Perrella
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Caroline Kardeby
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Magdolna Nagy
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Diego Mezzano
- Laboratorio de Trombosis y Hemostasia, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Paula M Mendes
- School of Chemical Engineering, College of Engineering and Physical Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, UK
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, UK
| |
Collapse
|
25
|
Clark JC, Damaskinaki FN, Cheung YFH, Slater A, Watson SP. Structure-function relationship of the platelet glycoprotein VI (GPVI) receptor: does it matter if it is a dimer or monomer? Platelets 2021; 32:724-732. [PMID: 33634725 DOI: 10.1080/09537104.2021.1887469] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/20/2020] [Accepted: 10/26/2020] [Indexed: 10/22/2022]
Abstract
GPVI is a critical signaling receptor responsible for collagen-induced platelet activation and a promising anti-thrombotic target in conditions such as coronary artery thrombosis, ischemic stroke, and atherothrombosis. This is due to the ability to block GPVI while having minimal effects on hemostasis, making it a more attractive target over current dual-antiplatelet therapy (DAPT) with acetyl salicylic acid and P2Y12 inhibitors where bleeding can be a problem. Our current understanding of how the structure of GPVI relates to function is inadequate and recent studies contradict each other. In this article, we summarize the structure-function relationships underlying the activation of GPVI by its major ligands, including collagen, fibrin(ogen), snake venom toxins and charged exogenous ligands such as diesel exhaust particles. We argue that contrary to popular belief dimerization of GPVI is not required for binding to collagen but serves to facilitate binding through increased avidity, and that GPVI is expressed as a mixture of monomers and dimers on resting platelets, with binding of multivalent ligands inducing higher order clustering.
Collapse
Affiliation(s)
- Joanne C Clark
- 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
| | - 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
- School of Pharmacy, Biodiscovery Institute, University Park, University of Nottingham, Nottingham, UK
| | - Yam Fung Hilaire Cheung
- Institute of Cardiovascular Sciences, Level 1 IBR, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Department of Bioanalytics, Leibniz-Institut Für Analytische Wissenschaften - ISAS -e.v, Dortmund, Germany
| | - Alexandre Slater
- Institute of Cardiovascular Sciences, Level 1 IBR, College of Medical and Dental Sciences, University of Birmingham, Birmingham, 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
|
26
|
Gu X, Zhang J, Li J, Wang Z, Feng J, Li J, Pan K, Ni X, Zeng D, Jing B, Zhang D. Effects of Bacillus cereus PAS38 on Immune-Related Differentially Expressed Genes of Spleen in Broilers. Probiotics Antimicrob Proteins 2021; 12:425-438. [PMID: 31243733 DOI: 10.1007/s12602-019-09567-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This study mainly explored the immunomodulatory mechanisms of the probiotic Bacillus cereus PAS38 (PB) on broiler spleen. A total of 120 avian white feather broilers were randomly divided into 4 groups (N = 30), as follows: control (CNTL, fed with basal diet), PB (fed with diet supplemented with probiotic B. cereus PAS38), vaccine (VAC, fed with basal diet and injected with Newcastle disease virus vaccine), and vaccine + PB group (PBVAC, fed with basal diet supplemented with B. cereus PAS38 and injected with NDV vaccine). The experiment was conducted for 42 days. Twelve spleens were collected from four different groups, weighed, and cut into histological sections, and transcriptome analysis was performed using RNA-seq. Results of the spleen and histological section relative weights showed that feeding with probiotic B. cereus PAS38 and vaccination had a similar tendency to promote spleen development. Compared with the CNTL group, 21 immune-related genes were significantly downregulated in the PB and PBVAC groups. These genes were mainly involved in attenuating inflammatory response. The upregulated antimicrobial peptide NK-lysin and guanylate-binding protein 1 expression levels indicated that this strain enhanced the body's antimicrobial capacity. B. cereus PAS38 also amplified the broilers' immune response to the vaccine, which mainly reflected on nonspecific immunity. Hence, probiotic B. cereus PAS38 can regulate and promote the immune function of broilers.
Collapse
Affiliation(s)
- Xiaoxiao Gu
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Jiao Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Jiajun Li
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Zhenhua Wang
- Chengdu Vocational College of Agricultural Science and Technology, Chengdu, 611100, China
| | - Jie Feng
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Jianzhen Li
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
- Chengdu Vocational College of Agricultural Science and Technology, Chengdu, 611100, China
| | - Kangcheng Pan
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China.
| | - Xueqin Ni
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Dong Zeng
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Bo Jing
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| | - Dongmei Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wengjiang District, Chengdu, 611130, Sichuan, People's Republic of China
| |
Collapse
|
27
|
Structural characterization of a novel GPVI-nanobody complex reveals a biologically active domain-swapped GPVI dimer. Blood 2021; 137:3443-3453. [PMID: 33512486 DOI: 10.1182/blood.2020009440] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/14/2021] [Indexed: 12/22/2022] Open
Abstract
Glycoprotein VI (GPVI) is the major signaling receptor for collagen on platelets. We have raised 54 nanobodies (Nb), grouped into 33 structural classes based on their complementary determining region 3 loops, against recombinant GPVI-Fc (dimeric GPVI) and have characterized their ability to bind recombinant GPVI, resting and activated platelets, and to inhibit platelet activation by collagen. Nbs from 6 different binding classes showed the strongest binding to recombinant GPVI-Fc, suggesting that there was not a single dominant class. The most potent 3, Nb2, 21, and 35, inhibited collagen-induced platelet aggregation with nanomolar half maximal inhibitory concentration (IC50) values and inhibited platelet aggregation under flow. The binding KD of the most potent Nb, Nb2, against recombinant monomeric and dimeric GPVI was 0.6 and 0.7 nM, respectively. The crystal structure of monomeric GPVI in complex with Nb2 revealed a binding epitope adjacent to the collagen-related peptide (CRP) binding groove within the D1 domain. In addition, a novel conformation of GPVI involving a domain swap between the D2 domains was observed. The domain swap is facilitated by the outward extension of the C-C' loop, which forms the domain swap hinge. The functional significance of this conformation was tested by truncating the hinge region so that the domain swap cannot occur. Nb2 was still able to displace collagen and CRP binding to the mutant, but signaling was abolished in a cell-based NFAT reporter assay. This demonstrates that the C-C' loop region is important for GPVI signaling but not ligand binding and suggests the domain-swapped structure may represent an active GPVI conformation.
Collapse
|
28
|
Clark JC, Neagoe RAI, Zuidscherwoude M, Kavanagh DM, Slater A, Martin EM, Soave M, Stegner D, Nieswandt B, Poulter NS, Hummert J, Herten DP, Tomlinson MG, Hill SJ, Watson SP. Evidence that GPVI is Expressed as a Mixture of Monomers and Dimers, and that the D2 Domain is not Essential for GPVI Activation. Thromb Haemost 2021; 121:1435-1447. [PMID: 33638140 DOI: 10.1055/a-1401-5014] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Collagen has been proposed to bind to a unique epitope in dimeric glycoprotein VI (GPVI) and the number of GPVI dimers has been reported to increase upon platelet activation. However, in contrast, the crystal structure of GPVI in complex with collagen-related peptide (CRP) showed binding distinct from the site of dimerization. Further fibrinogen has been reported to bind to monomeric but not dimeric GPVI. In the present study, we have used the advanced fluorescence microscopy techniques of single-molecule microscopy, fluorescence correlation spectroscopy (FCS) and bioluminescence resonance energy transfer (BRET), and mutagenesis studies in a transfected cell line model to show that GPVI is expressed as a mixture of monomers and dimers and that dimerization through the D2 domain is not critical for activation. As many of these techniques cannot be applied to platelets to resolve this issue, due to the high density of GPVI and its anucleate nature, we used Förster resonance energy transfer (FRET) to show that endogenous GPVI is at least partially expressed as a dimer on resting and activated platelet membranes. We propose that GPVI may be expressed as a monomer on the cell surface and it forms dimers in the membrane through diffusion, giving rise to a mixture of monomers and dimers. We speculate that the formation of dimers facilitates ligand binding through avidity.
Collapse
Affiliation(s)
- Joanne C Clark
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors, The Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| | - Raluca A I Neagoe
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Institute of Experimental Biomedicine I, University Hospital and Rudolf Virchow Center, University of Wurzburg, Wurzburg, Germany
| | - Malou Zuidscherwoude
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors, The Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| | - Deirdre M Kavanagh
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors, The Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| | - Alexandre Slater
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Eleyna M Martin
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Mark Soave
- Centre of Membrane Proteins and Receptors, The Universities of Birmingham and Nottingham, The Midlands, United Kingdom.,Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - David Stegner
- Institute of Experimental Biomedicine I, University Hospital and Rudolf Virchow Center, University of Wurzburg, Wurzburg, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine I, University Hospital and Rudolf Virchow Center, University of Wurzburg, Wurzburg, Germany
| | - Natalie S Poulter
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors, The Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| | - Johan Hummert
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors, The Universities of Birmingham and Nottingham, The Midlands, United Kingdom.,Department for Physical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Dirk-Peter Herten
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors, The Universities of Birmingham and Nottingham, The Midlands, United Kingdom.,Department for Physical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Michael G Tomlinson
- Centre of Membrane Proteins and Receptors, The Universities of Birmingham and Nottingham, The Midlands, United Kingdom.,School of Biosciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Stephen J Hill
- Centre of Membrane Proteins and Receptors, The Universities of Birmingham and Nottingham, The Midlands, United Kingdom.,Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors, The Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| |
Collapse
|
29
|
Xu RG, Gauer JS, Baker SR, Slater A, Martin EM, McPherson HR, Duval C, Manfield IW, Bonna AM, Watson SP, Ariëns RAS. GPVI (Glycoprotein VI) Interaction With Fibrinogen Is Mediated by Avidity and the Fibrinogen αC-Region. Arterioscler Thromb Vasc Biol 2021; 41:1092-1104. [PMID: 33472402 PMCID: PMC7901536 DOI: 10.1161/atvbaha.120.315030] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. Objective: GPVI (glycoprotein VI) is a key molecular player in collagen-induced platelet signaling and aggregation. Recent evidence indicates that it also plays important role in platelet aggregation and thrombus growth through interaction with fibrin(ogen). However, there are discrepancies in the literature regarding whether the monomeric or dimeric form of GPVI binds to fibrinogen at high affinity. The mechanisms of interaction are also not clear, including which region of fibrinogen is responsible for GPVI binding. We aimed to gain further understanding of the mechanisms of interaction at molecular level and to identify the regions on fibrinogen important for GPVI binding. Approach and Results: Using multiple surface- and solution-based protein-protein interaction methods, we observe that dimeric GPVI binds to fibrinogen with much higher affinity and has a slower dissociation rate constant than the monomer due to avidity effects. Moreover, our data show that the highest affinity interaction of GPVI is with the αC-region of fibrinogen. We further show that GPVI interacts with immobilized fibrinogen and fibrin variants at a similar level, including a nonpolymerizing fibrin variant, suggesting that GPVI binding is independent of fibrin polymerization. Conclusions: Based on the above findings, we conclude that the higher affinity of dimeric GPVI over the monomer for fibrinogen interaction is achieved by avidity. The αC-region of fibrinogen appears essential for GPVI binding. We propose that fibrin polymerization into fibers during coagulation will cluster GPVI through its αC-region, leading to downstream signaling, further activation of platelets, and potentially stimulating clot growth.
Collapse
Affiliation(s)
- Rui-Gang Xu
- Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine (R.-G.X., J.S.G., S.R.B., H.R.M., C.D., R.A.S.A.)
| | - Julia S Gauer
- Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine (R.-G.X., J.S.G., S.R.B., H.R.M., C.D., R.A.S.A.)
| | - Stephen R Baker
- Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine (R.-G.X., J.S.G., S.R.B., H.R.M., C.D., R.A.S.A.).,Department of Physics, Wake Forest University, Winston Salem, NC (S.R.B.)
| | - Alexandre Slater
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (A.S., E.M.M., S.P.W.)
| | - Eleyna M Martin
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (A.S., E.M.M., S.P.W.)
| | - Helen R McPherson
- Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine (R.-G.X., J.S.G., S.R.B., H.R.M., C.D., R.A.S.A.)
| | - Cédric Duval
- Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine (R.-G.X., J.S.G., S.R.B., H.R.M., C.D., R.A.S.A.)
| | - Iain W Manfield
- School of Molecular and Cellular Biology, Faculty of Biological Sciences (I.W.M.), University of Leeds, United Kingdom
| | - Arkadiusz M Bonna
- Department of Biochemistry, University of Cambridge, United Kingdom (A.M.B.)
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (A.S., E.M.M., S.P.W.)
| | - Robert A S Ariëns
- Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine (R.-G.X., J.S.G., S.R.B., H.R.M., C.D., R.A.S.A.)
| |
Collapse
|
30
|
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: 13] [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
|
31
|
Borst O, Gawaz M. Glycoprotein VI - novel target in antiplatelet medication. Pharmacol Ther 2021; 217:107630. [DOI: 10.1016/j.pharmthera.2020.107630] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/11/2020] [Indexed: 02/07/2023]
|
32
|
Collagen Structure-Function Mapping Informs Applications for Regenerative Medicine. Bioengineering (Basel) 2020; 8:bioengineering8010003. [PMID: 33383610 PMCID: PMC7824244 DOI: 10.3390/bioengineering8010003] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/21/2022] Open
Abstract
Type I collagen, the predominant protein of vertebrates, assembles into fibrils that orchestrate the form and function of bone, tendon, skin, and other tissues. Collagen plays roles in hemostasis, wound healing, angiogenesis, and biomineralization, and its dysfunction contributes to fibrosis, atherosclerosis, cancer metastasis, and brittle bone disease. To elucidate the type I collagen structure-function relationship, we constructed a type I collagen fibril interactome, including its functional sites and disease-associated mutations. When projected onto an X-ray diffraction model of the native collagen microfibril, data revealed a matrix interaction domain that assumes structural roles including collagen assembly, crosslinking, proteoglycan (PG) binding, and mineralization, and the cell interaction domain supporting dynamic aspects of collagen biology such as hemostasis, tissue remodeling, and cell adhesion. Our type III collagen interactome corroborates this model. We propose that in quiescent tissues, the fibril projects a structural face; however, tissue injury releases blood into the collagenous stroma, triggering exposure of the fibrils' cell and ligand binding sites crucial for tissue remodeling and regeneration. Applications of our research include discovery of anti-fibrotic antibodies and elucidating their interactions with collagen, and using insights from our angiogenesis studies and collagen structure-function model to inform the design of super-angiogenic collagens and collagen mimetics.
Collapse
|
33
|
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: 0.8] [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
|
34
|
Abstract
Acute coronary syndromes (ACS) are a global cause of mortality and morbidity that affect millions of lives worldwide. Following atherosclerotic plaque rupture, platelet activation and aggregation are the two major elements that initiate thrombus formation inside a coronary artery, which can obstruct blood flow and cause myocardial ischemia; ergo, antiplatelet therapy forms a major part of the treatment strategy for ACS. Patients with ACS routinely receive dual antiplatelet therapy (DAPT), which consists of aspirin and a platelet P2Y12 inhibitor to both treat and prevent atherothrombosis. Use of platelet glycoprotein (GP) IIb/IIIa inhibitors is now limited due to the risk of severe bleeding and thrombocytopenia. Thus, administration of GPIIb/IIIa inhibitors is generally restricted to bail out thrombotic events associated with PCI. Furthermore, current antiplatelet medications mainly rely on thromboxane A2 and P2Y12 inhibition, which have broad-acting effects on platelets and are known to cause bleeding, which especially limits the long-term use of these agents. In addition, not all ACS patients treated with current antiplatelet treatments are protected from recurrence of arterial thrombosis, since many platelet mechanisms and activation pathways remain uninhibited by current antiplatelet therapy. Pharmacological antagonism of novel targets involved in platelet function could shape future antiplatelet therapies that could ultimately lead to more effective or safer therapeutic approaches. In this article, we focus on inhibitors of promising targets that have not yet been introduced into clinical practice, including inhibitors of GPVI, protease-activated receptor (PAR)-4, GPIb, 5-hydroxytryptamine receptor subtype 2A (5-HT2A), protein disulfide isomerase, P-selectin and phosphoinositide 3-kinase β.
Collapse
Affiliation(s)
- Fawaz O Alenazy
- Institute of Cardiovascular Sciences, University of Birmingham , Birmingham, UK
| | - Mark R Thomas
- Institute of Cardiovascular Sciences, University of Birmingham , Birmingham, UK.,UHB and SWBH NHS Trusts , Birmingham, UK
| |
Collapse
|
35
|
Zhu J, Madhurapantula RS, Kalyanasundaram A, Sabharwal T, Antipova O, Bishnoi SW, Orgel JPRO. Ultrastructural Location and Interactions of the Immunoglobulin Receptor Binding Sequence within Fibrillar Type I Collagen. Int J Mol Sci 2020; 21:ijms21114166. [PMID: 32545195 PMCID: PMC7312686 DOI: 10.3390/ijms21114166] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 12/30/2022] Open
Abstract
Collagen type I is a major constituent of animal bodies. It is found in large quantities in tendon, bone, skin, cartilage, blood vessels, bronchi, and the lung interstitium. It is also produced and accumulates in large amounts in response to certain inflammations such as lung fibrosis. Our understanding of the molecular organization of fibrillar collagen and cellular interaction motifs, such as those involved with immune-associated molecules, continues to be refined. In this study, antibodies raised against type I collagen were used to label intact D-periodic type I collagen fibrils and observed with atomic force microscopy (AFM), and X-ray diffraction (XRD) and immunolabeling positions were observed with both methods. The antibodies bind close to the C-terminal telopeptide which verifies the location and accessibility of both the major histocompatibility complex (MHC) class I (MHCI) binding domain and C-terminal telopeptide on the outside of the collagen fibril. The close proximity of the C-telopeptide and the MHC1 domain of type I collagen to fibronectin, discoidin domain receptor (DDR), and collagenase cleavage domains likely facilitate the interaction of ligands and receptors related to cellular immunity and the collagen-based Extracellular Matrix.
Collapse
Affiliation(s)
- Jie Zhu
- Institute of Biophysics, College of science, Northwest A&F University, Yangling 712100, China
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA; (R.S.M.); (A.K.); (T.S.); (O.A.)
- Pritzker Institute of Biomedical Science and Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA
- Correspondence: (J.Z.); (J.P.R.O.O.)
| | - Rama S. Madhurapantula
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA; (R.S.M.); (A.K.); (T.S.); (O.A.)
- Pritzker Institute of Biomedical Science and Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Aruna Kalyanasundaram
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA; (R.S.M.); (A.K.); (T.S.); (O.A.)
| | - Tanya Sabharwal
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA; (R.S.M.); (A.K.); (T.S.); (O.A.)
| | - Olga Antipova
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA; (R.S.M.); (A.K.); (T.S.); (O.A.)
- X-ray Science Division, Argonne National Laboratory, Lemont, IL 60439, USA
| | - Sandra W. Bishnoi
- Department of Chemistry, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Joseph P. R. O. Orgel
- Department of Biology, Illinois Institute of Technology, Chicago, IL 60616, USA; (R.S.M.); (A.K.); (T.S.); (O.A.)
- Pritzker Institute of Biomedical Science and Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA
- Correspondence: (J.Z.); (J.P.R.O.O.)
| |
Collapse
|
36
|
Montague SJ, Hicks SM, Lee CSM, Coupland LA, Parish CR, Lee WM, Andrews RK, Gardiner EE. Fibrin exposure triggers αIIbβ3-independent platelet aggregate formation, ADAM10 activity and glycoprotein VI shedding in a charge-dependent manner. J Thromb Haemost 2020; 18:1447-1458. [PMID: 32198957 DOI: 10.1111/jth.14797] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 03/11/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Collagen and fibrin engagement and activation of glycoprotein (GP) VI induces proteolytic cleavage of the GPVI ectodomain generating shed soluble GPVI (sGPVI). Collagen-mediated GPVI shedding requires intracellular signalling to release the sGPVI, mediated by A Disintegrin And Metalloproteinase 10 (ADAM10); however, the precise mechanism by which fibrin induces GPVI shedding remains elusive. Plasma sGPVI levels are elevated in patients with coagulopathies, sepsis, or inflammation and can predict onset of sepsis and sepsis-related mortality; therefore, it is clinically important to understand the mechanisms of GPVI shedding under conditions of minimal collagen exposure. OBJECTIVES Our aim was to characterize mechanisms by which fibrin-GPVI interactions trigger GPVI shedding. METHODS Platelet aggregometry, sGPVI ELISA, and an ADAM10 fluorescence resonance energy transfer assay were used to measure fibrin-mediated platelet responses. RESULTS Fibrin induced αIIbβ3-independent washed platelet aggregate formation, GPVI shedding, and increased ADAM10 activity, all of which were insensitive to pre-treatment with inhibitors of Src family kinases but were divalent cation- and metalloproteinase-dependent. In contrast, treatment of washed platelets with other GPVI ligands, collagen, and collagen-related peptide caused αIIbβ3-dependent platelet aggregation and GPVI release but did not increase constitutive ADAM10 activity. CONCLUSIONS Fibrin engages GPVI in a manner that differs from other GPVI ligands. Inclusion of polyanionic molecules disrupted fibrin-induced platelet aggregate formation and sGPVI release, suggesting that electrostatic charge may play a role in fibrin/GPVI engagement. It may be feasible to exploit this property and specifically disrupt GPVI/fibrin interactions whilst sparing GPVI/collagen engagement.Fibrin engages GPVI in a manner that differs from other GPVI ligands. Inclusion of polyanionic molecules disrupted fibrin-induced platelet aggregate formation and sGPVI release, suggesting that electrostatic charge may play a role in fibrin/GPVI engagement. It may be feasible to exploit this property and specifically disrupt GPVI/fibrin interactions whilst sparing GPVI/collagen engagement.
Collapse
Affiliation(s)
- Samantha J Montague
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Sarah M Hicks
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Christine S-M Lee
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Lucy A Coupland
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Christopher R Parish
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Woei M Lee
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
- Research School of Electrical, Energy and Materials Engineering, College of Engineering and Computer Science, The Australian National University, Canberra, ACT, Australia
| | - Robert K Andrews
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Elizabeth E Gardiner
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
37
|
Hoop CL, Zhu J, Bhattacharya S, Tobita CA, Radford SE, Baum J. Collagen I Weakly Interacts with the β-Sheets of β 2-Microglobulin and Enhances Conformational Exchange To Induce Amyloid Formation. J Am Chem Soc 2020; 142:1321-1331. [PMID: 31875390 PMCID: PMC7135851 DOI: 10.1021/jacs.9b10421] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
![]()
Amyloidogenesis is
significant in both protein function and pathology.
Amyloid formation of folded, globular proteins is commonly initiated
by partial or complete unfolding. However, how this unfolding event
is triggered for proteins that are otherwise stable in their native
environments is not well understood. The accumulation of the immunoglobulin
protein β2-microglobulin (β2m) into
amyloid plaques in the joints of long-term hemodialysis patients is
the hallmark of dialysis-related amyloidosis (DRA). While β2m does not form amyloid unassisted near neutral pH in vitro, the localization of β2m deposits
to joint spaces suggests a role for the local extracellular matrix
(ECM) proteins, specifically collagens, in promoting amyloid formation.
Indeed, collagen and other ECM components have been observed to facilitate
β2m amyloid formation, but the large size and anisotropy
of the complex, combined with the low affinity of these interactions,
have limited atomic-level elucidation of the amyloid-promoting mechanism(s)
by these molecules. Using solution NMR approaches that uniquely probe
weak interactions in large molecular weight complexes, we are able
to map the binding interfaces on β2m for collagen
I and detect collagen I-induced μs–ms time-scale dynamics
in the β2m backbone. By combining solution NMR relaxation
methods and 15N-dark-state exchange saturation transfer
experiments, we propose a model in which weak, multimodal collagen
I−β2m interactions promote exchange with a
minor population of amyloid-competent species to induce fibrillogenesis.
The results portray the intimate role of the environment in switching
an innocuous protein into an amyloid-competent state, rationalizing
the localization of amyloid deposits in DRA.
Collapse
Affiliation(s)
- Cody L Hoop
- Department of Chemistry and Chemical Biology , Rutgers University , Piscataway , New Jersey 08854 , United States
| | - Jie Zhu
- Department of Chemistry and Chemical Biology , Rutgers University , Piscataway , New Jersey 08854 , United States
| | | | - Caitlyn A Tobita
- Department of Chemistry and Chemical Biology , Rutgers University , Piscataway , New Jersey 08854 , United States
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology and School of Molecular and Cellular Biology, Faculty of Biological Sciences , University of Leeds , Leeds LS2 9JT , U.K
| | - Jean Baum
- Department of Chemistry and Chemical Biology , Rutgers University , Piscataway , New Jersey 08854 , United States
| |
Collapse
|
38
|
Jandrot-Perrus M, Hermans C, Mezzano D. Platelet glycoprotein VI genetic quantitative and qualitative defects. Platelets 2019; 30:708-713. [PMID: 31068042 DOI: 10.1080/09537104.2019.1610166] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Platelet membrane glycoprotein VI (GPVI) is increasingly recognized as an important receptor for thrombus formation and growth. Numerous arguments have been published indicating that GPVI plays a major role in thrombosis without being essential for physiological hemostasis. In humans, GPVI deficiencies are rarely reported. These are most often deficiencies occurring in the context of autoimmunity and, more rarely, genetic deficits. The purpose of this review is to compile data on the quantitative and qualitative genetic abnormalities of GPVI.
Collapse
Affiliation(s)
- Martine Jandrot-Perrus
- a UMR_S1148, Laboratory for Vascular Translational Science , INSERM, University Paris Diderot , Paris , France
| | - Cedric Hermans
- b Haemostasis and Thrombosis Unit, Haemophilia Clinic, Division of Adult Haematology , St-Luc University Hospital , Brussels , Belgium
| | - Diego Mezzano
- c Department of Hematology-Oncology , School of Medicine, Pontificia Universidad Católica de Chile , Santiago , Chile
| |
Collapse
|
39
|
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: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 02/12/2019] [Indexed: 01/04/2023]
|
40
|
Orgel JPRO, Madhurapantula RS. A structural prospective for collagen receptors such as DDR and their binding of the collagen fibril. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:118478. [PMID: 31004686 DOI: 10.1016/j.bbamcr.2019.04.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 04/11/2019] [Accepted: 04/12/2019] [Indexed: 12/13/2022]
Abstract
The structure of the collagen fibril surface directly effects and possibly assists the management of collagen receptor interactions. An important class of collagen receptors, the receptor tyrosine kinases of the Discoidin Domain Receptor family (DDR1 and DDR2), are differentially activated by specific collagen types and play important roles in cell adhesion, migration, proliferation, and matrix remodeling. This review discusses their structure and function as it pertains directly to the fibrillar collagen structure with which they interact far more readily than they do with isolated molecular collagen. This prospective provides further insight into the mechanisms of activation and rational cellular control of this important class of receptors while also providing a comparison of DDR-collagen interactions with other receptors such as integrin and GPVI. When improperly regulated, DDR activation can lead to abnormal cellular proliferation activities such as in cancer. Hence how and when the DDRs associate with the major basis of mammalian tissue infrastructure, fibrillar collagen, should be of keen interest.
Collapse
Affiliation(s)
- Joseph P R O Orgel
- Departments of Biology and Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA.
| | - Rama S Madhurapantula
- Departments of Biology and Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| |
Collapse
|
41
|
Rayes J, Watson SP, Nieswandt B. Functional significance of the platelet immune receptors GPVI and CLEC-2. J Clin Invest 2019; 129:12-23. [PMID: 30601137 DOI: 10.1172/jci122955] [Citation(s) in RCA: 217] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Although platelets are best known for their role in hemostasis, they are also crucial in development, host defense, inflammation, and tissue repair. Many of these roles are regulated by the immune-like receptors glycoprotein VI (GPVI) and C-type lectin receptor 2 (CLEC-2), which signal through an immunoreceptor tyrosine-based activation motif (ITAM). GPVI is activated by collagen in the subendothelial matrix, by fibrin and fibrinogen in the thrombus, and by a remarkable number of other ligands. CLEC-2 is activated by the transmembrane protein podoplanin, which is found outside of the vasculature and is upregulated in development, inflammation, and cancer, but there is also evidence for additional ligands. In this Review, we discuss the physiological and pathological roles of CLEC-2 and GPVI and their potential as targets in thrombosis and thrombo-inflammatory disorders (i.e., disorders in which inflammation plays a critical role in the ensuing thrombosis) relative to current antiplatelet drugs.
Collapse
Affiliation(s)
- Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, United Kingdom
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| |
Collapse
|
42
|
|
43
|
Saelao P, Wang Y, Gallardo RA, Lamont SJ, Dekkers JM, Kelly T, Zhou H. Novel insights into the host immune response of chicken Harderian gland tissue during Newcastle disease virus infection and heat treatment. BMC Vet Res 2018; 14:280. [PMID: 30208883 PMCID: PMC6134752 DOI: 10.1186/s12917-018-1583-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/17/2018] [Indexed: 01/05/2023] Open
Abstract
Background Newcastle disease virus, in its most pathogenic form, threatens the livelihood of rural poultry farmers where there is a limited infrastructure and service for vaccinations to prevent outbreaks of the virus. Previously reported studies on the host response to Newcastle disease in chickens have not examined the disease under abiotic stressors, such as heat, which commonly experienced by chickens in regions such as Africa. The objective of this study was to elucidate the underlying biological mechanisms that contribute to disease resistance in chickens to the Newcastle disease virus while under the effects of heat stress. Results Differential gene expression analysis identified genes differentially expressed between treated and non-treated birds across three time points (2, 6, and 10 days post-infection) in Fayoumi and Leghorn birds. Across the three time points, Fayoumi had very few genes differentially expressed between treated and non-treated groups at 2 and 6 days post-infection. However, 202 genes were differentially expressed at 10 days post-infection. Alternatively, Leghorn had very few genes differentially expressed at 2 and 10 days post-infection but had 167 differentially expressed genes at 6 days post-infection. Very few differentially expressed genes were shared between the two genetic lines, and pathway analysis found unique signaling pathways specific to each genetic line. Fayoumi had significantly lower viral load, higher viral clearance, higher anti-NDV antibody levels, and fewer viral transcripts detected compared to Leghorns. Fayoumis activated immune related pathways including SAPK/JNK and p38 MAPK signaling pathways at earlier time points, while Leghorn would activate these same pathways at a later time. Further analysis revealed activation of the GP6 signaling pathway that may be responsible for the susceptible Leghorn response. Conclusions The findings in this study confirmed our hypothesis that the Fayoumi line was more resistant to Newcastle disease virus infection compared to the Leghorn line. Within line and interaction analysis demonstrated substantial differences in response patterns between the two genetic lines that was not observed from the within line contrasts. This study has provided novel insights into the transcriptome response of the Harderian gland tissue during Newcastle disease virus infection while under heat stress utilizing a unique resistant and susceptible model. Electronic supplementary material The online version of this article (10.1186/s12917-018-1583-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Perot Saelao
- Integrative Genetics and Genomics Graduate Group, University of California, Davis, CA, 95616, USA.,Genomics to Improve Poultry Innovation Lab, University of California, Davis, CA, 95616, USA.,Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Ying Wang
- Genomics to Improve Poultry Innovation Lab, University of California, Davis, CA, 95616, USA.,Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Rodrigo A Gallardo
- School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Susan J Lamont
- Department of Animal Science, Iowa State University, Ames, IA, 50011, USA
| | - Jack M Dekkers
- Department of Animal Science, Iowa State University, Ames, IA, 50011, USA
| | - Terra Kelly
- Genomics to Improve Poultry Innovation Lab, University of California, Davis, CA, 95616, USA.,One Health Institute, University of California, Davis, CA, 95616, USA
| | - Huaijun Zhou
- Genomics to Improve Poultry Innovation Lab, University of California, Davis, CA, 95616, USA. .,Department of Animal Science, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
44
|
Slater A, Perrella G, Onselaer MB, Martin EM, Gauer JS, Xu RG, Heemskerk JWM, Ariëns RAS, Watson SP. Does fibrin(ogen) bind to monomeric or dimeric GPVI, or not at all? Platelets 2018; 30:281-289. [DOI: 10.1080/09537104.2018.1508649] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Alexandre Slater
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Gina Perrella
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands
| | - Marie-Blanche Onselaer
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Eleyna M Martin
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Julia S Gauer
- Thrombosis and Tissue Repair Group, Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Rui-Gang Xu
- Thrombosis and Tissue Repair Group, Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Johan WM Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands
| | - Robert A S Ariëns
- Thrombosis and Tissue Repair Group, Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
45
|
Howes J, Pugh N, Hamaia SW, Jung SM, Knäuper V, Malcor J, Farndale RW. MMP-13 binds to platelet receptors αIIbβ3 and GPVI and impairs aggregation and thrombus formation. Res Pract Thromb Haemost 2018; 2:370-379. [PMID: 30046741 PMCID: PMC5974921 DOI: 10.1002/rth2.12088] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 01/28/2018] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Acute thrombotic syndromes lead to atherosclerotic plaque rupture with subsequent thrombus formation, myocardial infarction and stroke. Following rupture, flowing blood is exposed to plaque components, including collagen, which triggers platelet activation and aggregation. However, plaque rupture releases other components into the surrounding vessel which have the potential to influence platelet function and thrombus formation. OBJECTIVES Here we sought to elucidate whether matrix metalloproteinase-13 (MMP-13), a collagenolytic metalloproteinase up-regulated in atherothrombotic and inflammatory conditions, affects platelet aggregation and thrombus formation. RESULTS We demonstrate that MMP-13 is able to bind to platelet receptors alphaIIbbeta3 (αIIbβ3) and platelet glycoprotein (GP)VI. The interactions between MMP-13, GPVI and αIIbβ3 are sufficient to significantly inhibit washed platelet aggregation and decrease thrombus formation on fibrillar collagen. CONCLUSIONS Our data demonstrate a role for MMP-13 in the inhibition of both platelet aggregation and thrombus formation in whole flowing blood, and may provide new avenues of research into the mechanisms underlying the subtle role of MMP-13 in atherothrombotic pathologies.
Collapse
Affiliation(s)
| | - Nicholas Pugh
- Department of Biomedical and Forensic SciencesAnglia Ruskin UniversityCambridgeUK
| | - Samir W. Hamaia
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | | | | | | | | |
Collapse
|
46
|
Chen J, Wang B, Wu Y. Structural Characterization and Function Prediction of Immunoglobulin-like Fold in Cell Adhesion and Cell Signaling. J Chem Inf Model 2018; 58:532-542. [PMID: 29356528 DOI: 10.1021/acs.jcim.7b00580] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Domains that belong to an immunoglobulin (Ig) fold are extremely abundant in cell surface receptors, which play significant roles in cell-cell adhesion and signaling. Although the structures of domains in an Ig fold share common topology of β-barrels, functions of receptors in adhesion and signaling are regulated by the very heterogeneous binding between these domains. Additionally, only a small number of domains are directly involved in the binding between two multidomain receptors. It is challenging and time consuming to experimentally detect the binding partners of a given receptor and further determine which specific domains in this receptor are responsible for binding. Therefore, current knowledge in the binding mechanism of Ig-fold domains and their impacts on cell adhesion and signaling is very limited. A bioinformatics study can shed light on this topic from a systematic point of view. However, there is so far no computational analysis on the structural and functional characteristics of the entire Ig fold. We constructed nonredundant structural data sets for all domains in Ig fold, depending on their functions in cell adhesion and signaling. We found that data sets of domains in adhesion receptors show different binding preference from domains in signaling receptors. Using structural alignment, we further built a common structural template for each group of a domain data set. By mapping the protein-protein binding interface of each domain in a group onto the surface of its structural template, we found binding interfaces are highly overlapped within each specific group. These overlapped interfaces, we called consensus binding interfaces, are distinguishable among different data sets of domains. Finally, the residue compositions on the consensus interfaces were used as indicators for multiple machine learning algorithms to predict if they can form homotypic interactions with each other. The overall performance of the cross-validation tests shows that our prediction accuracies ranged between 0.6 and 0.8.
Collapse
Affiliation(s)
- Jiawen Chen
- Department of Systems and Computational Biology, Albert Einstein College of Medicine , 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Bo Wang
- Department of Systems and Computational Biology, Albert Einstein College of Medicine , 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Yinghao Wu
- Department of Systems and Computational Biology, Albert Einstein College of Medicine , 1300 Morris Park Avenue, Bronx, New York 10461, United States
| |
Collapse
|
47
|
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
|
48
|
Abstract
Fibrin has recently been shown to activate platelets through the immunoglobulin receptor glycoprotein VI (GPVI). In the present study, we show that spreading of human platelets on fibrin is abolished in patients deficient in GPVI, confirming that fibrin activates human platelets through the immunoglobulin receptor. Using a series of proteolytic fragments, we show that D-dimer, but not the E fragment of fibrin, binds to GPVI and that immobilized D-dimer induces platelet spreading through activation of Src and Syk tyrosine kinases. In contrast, when platelets are activated in suspension, soluble D-dimer inhibits platelet aggregation induced by fibrin and collagen, but not by a collagen-related peptide composed of a repeat GPO sequence or by thrombin. Using surface plasmon resonance, we demonstrate that fibrin binds selectively to monomeric GPVI with a KD of 302 nM, in contrast to collagen, which binds primarily to dimeric GPVI. These results establish GPVI as the major signaling receptor for fibrin in human platelets and provide evidence that fibrin binds to a distinct configuration of GPVI. This indicates that it may be possible to develop agents that selectively block the interaction of fibrin but not collagen with the immunoglobulin receptor. Such agents are required to establish whether selective targeting of either interaction has the potential to lead to development of an antithrombotic agent with a reduced effect on bleeding relative to current antiplatelet drugs.
Collapse
|
49
|
Affiliation(s)
- Andrew B. Herr
- From the Divisions of Immunobiology and Infectious Diseases, Cincinnati Children’s Hospital Medical Center, OH
| |
Collapse
|
50
|
Chang CH, Chung CH, Tu YS, Tsai CC, Hsu CC, Peng HC, Tseng YJ, Huang TF. Trowaglerix Venom Polypeptides As a Novel Antithrombotic Agent by Targeting Immunoglobulin-Like Domains of Glycoprotein VI in Platelet. Arterioscler Thromb Vasc Biol 2017; 37:1307-1314. [DOI: 10.1161/atvbaha.116.308604] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 04/25/2017] [Indexed: 11/16/2022]
Abstract
Objective—
Currently prescribed antiplatelet drugs have 1 common side effect—an increased risk of hemorrhage and thrombocytopenia. On the contrary, bleeding defects associated with glycoprotein VI (GPVI) expression deficiency are usually slightly prolonged bleeding times. However, GPVI antagonists are lacking in clinic.
Approach and Results—
Using reverse-phase high-performance liquid chromatography and sequencing, we revealed the partial sequence of trowaglerix α subunit, a potent specific GPVI-targeting snaclec (snake venom C-type lectin protein). Hexapeptide (Troα6 [trowaglerix a chain hexapeptide, CKWMNV]) and decapeptide (Troα10) derived from trowaglerix specifically inhibited collagen-induced platelet aggregation through blocking platelet GPVI receptor. Computational peptide design helped to design a series of Troα6/Troα10 peptides. Protein docking studies on these decapeptides and GPVI suggest that Troα10 was bound at the lower surface of D1 domain and outer surface of D2 domain, which was at the different place of the collagen-binding site and the scFv (single-chain variable fragment) D2-binding site. The newly discovered site was confirmed by inhibitory effects of polyclonal antibodies on collagen-induced platelet aggregation. This indicates that D2 domain of GPVI is a novel and important binding epitope on GPVI-mediated platelet aggregation. Troα6/Troα10 displayed prominent inhibitory effect of thrombus formation in fluorescein sodium-induced platelet thrombus formation of mesenteric venules and ferric chloride-induced carotid artery injury thrombosis model without prolonging the in vivo bleeding time.
Conclusions—
We develop a novel antithrombotic peptides derived from trowaglerix that acts through GPVI antagonism with greater safety—no severe bleeding. The binding epitope of polypeptides on GPVI is novel and important. These hexa/decapeptides have therapeutic potential for developing ideal small-mass GPVI antagonists for arterial thrombogenic diseases.
Collapse
Affiliation(s)
- Chien-Hsin Chang
- From the Graduate Institute of Pharmacology, College of Medicine (C.-H.C., C.-C.H., H.-C.P., T.-F.H.), Graduate Institute of Biomedical Electronics and Bioinformatics (Y.-S.T., C.-C.T., Y.J.T.), and Department of Computational Science and Information Engineering (Y.J.T.), National Taiwan University, Taipei; and Department of Medicine, Mackay Medical College, New Taipei City, Taiwan (C.-H.C.)
| | - Ching-Hu Chung
- From the Graduate Institute of Pharmacology, College of Medicine (C.-H.C., C.-C.H., H.-C.P., T.-F.H.), Graduate Institute of Biomedical Electronics and Bioinformatics (Y.-S.T., C.-C.T., Y.J.T.), and Department of Computational Science and Information Engineering (Y.J.T.), National Taiwan University, Taipei; and Department of Medicine, Mackay Medical College, New Taipei City, Taiwan (C.-H.C.)
| | - Yi-Shu Tu
- From the Graduate Institute of Pharmacology, College of Medicine (C.-H.C., C.-C.H., H.-C.P., T.-F.H.), Graduate Institute of Biomedical Electronics and Bioinformatics (Y.-S.T., C.-C.T., Y.J.T.), and Department of Computational Science and Information Engineering (Y.J.T.), National Taiwan University, Taipei; and Department of Medicine, Mackay Medical College, New Taipei City, Taiwan (C.-H.C.)
| | - Cheng-Chieh Tsai
- From the Graduate Institute of Pharmacology, College of Medicine (C.-H.C., C.-C.H., H.-C.P., T.-F.H.), Graduate Institute of Biomedical Electronics and Bioinformatics (Y.-S.T., C.-C.T., Y.J.T.), and Department of Computational Science and Information Engineering (Y.J.T.), National Taiwan University, Taipei; and Department of Medicine, Mackay Medical College, New Taipei City, Taiwan (C.-H.C.)
| | - Chun-Chieh Hsu
- From the Graduate Institute of Pharmacology, College of Medicine (C.-H.C., C.-C.H., H.-C.P., T.-F.H.), Graduate Institute of Biomedical Electronics and Bioinformatics (Y.-S.T., C.-C.T., Y.J.T.), and Department of Computational Science and Information Engineering (Y.J.T.), National Taiwan University, Taipei; and Department of Medicine, Mackay Medical College, New Taipei City, Taiwan (C.-H.C.)
| | - Hui-Chin Peng
- From the Graduate Institute of Pharmacology, College of Medicine (C.-H.C., C.-C.H., H.-C.P., T.-F.H.), Graduate Institute of Biomedical Electronics and Bioinformatics (Y.-S.T., C.-C.T., Y.J.T.), and Department of Computational Science and Information Engineering (Y.J.T.), National Taiwan University, Taipei; and Department of Medicine, Mackay Medical College, New Taipei City, Taiwan (C.-H.C.)
| | - Yufeng J. Tseng
- From the Graduate Institute of Pharmacology, College of Medicine (C.-H.C., C.-C.H., H.-C.P., T.-F.H.), Graduate Institute of Biomedical Electronics and Bioinformatics (Y.-S.T., C.-C.T., Y.J.T.), and Department of Computational Science and Information Engineering (Y.J.T.), National Taiwan University, Taipei; and Department of Medicine, Mackay Medical College, New Taipei City, Taiwan (C.-H.C.)
| | - Tur-Fu Huang
- From the Graduate Institute of Pharmacology, College of Medicine (C.-H.C., C.-C.H., H.-C.P., T.-F.H.), Graduate Institute of Biomedical Electronics and Bioinformatics (Y.-S.T., C.-C.T., Y.J.T.), and Department of Computational Science and Information Engineering (Y.J.T.), National Taiwan University, Taipei; and Department of Medicine, Mackay Medical College, New Taipei City, Taiwan (C.-H.C.)
| |
Collapse
|