1
|
Dangelmaier C, Vari HR, Vajipayajula DN, Elzoheiry M, Wright M, Iyer A, Tsygankov AY, Kunapuli SP. Phosphorylation of (Ser 291) in the linker insert of Syk negatively regulates ITAM signaling in platelets. Platelets 2024; 35:2369766. [PMID: 38904212 PMCID: PMC11322839 DOI: 10.1080/09537104.2024.2369766] [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] [Indexed: 06/22/2024]
Abstract
Receptor-induced tyrosine phosphorylation of spleen tyrosine kinase (Syk) has been studied extensively in hematopoietic cells. Metabolic mapping and high-resolution mass spectrometry, however, indicate that one of the most frequently detected phosphorylation sites encompassed S297 (S291 in mice) located within the linker B region of Syk. It has been reported that Protein kinase C (PKC) phosphorylates Syk S297, thus influencing Syk activity. However, conflicting studies suggest that this phosphorylation enhances as well as reduces Syk activity. To clarify the function of this site, we generated Syk S291A knock-in mice. We used platelets as a model system as they possess Glycoprotein VI (GPVI), a receptor containing an immunoreceptor tyrosine-based activation motif (ITAM) which transduces signals through Syk. Our analysis of the homozygous mice indicated that the knock-in platelets express only one isoform of Syk, while the wild-type expresses two isoforms at 69 and 66 kDa. When the GPVI receptor was activated with collagen-related peptide (CRP), we observed an increase in functional responses and phosphorylations in Syk S291A platelets. This potentiation did not occur with AYPGKF or 2-MeSADP, although they also activate PKC isoforms. Although there was potentiation of platelet functional responses, there was no difference in tail bleeding times. However, the time to occlusion in the FeCl3 injury model was enhanced. These data indicate that the effects of Syk S291 phosphorylation represent a significant outcome on platelet activation and signaling in vitro but also reveals its multifaceted nature demonstrated by the differential effects on physiological responses in vivo.
Collapse
Affiliation(s)
- Carol Dangelmaier
- Sol Sherry Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Hymavathi Reddy Vari
- Sol Sherry Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Dhruv N Vajipayajula
- Sol Sherry Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Manal Elzoheiry
- Sol Sherry Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Monica Wright
- Sol Sherry Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Ashvin Iyer
- Sol Sherry Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Alexander Y Tsygankov
- Sol Sherry Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Satya P Kunapuli
- Sol Sherry Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
2
|
Ye Y, Leng M, Chai S, Yang L, Ren L, Wan W, Wang H, Li L, Li C, Meng Z. Antiplatelet effects of the CEACAM1-derived peptide QDTT. Platelets 2024; 35:2308635. [PMID: 38345065 DOI: 10.1080/09537104.2024.2308635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/17/2024] [Indexed: 02/15/2024]
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) restricts platelet activation via platelet collagen receptor GPVI/FcRγ-chain. In this study, screening against collagen-induced platelet aggregation was performed to identify functional CEACAM1 extracellular domain fragments. CEACAM1 fragments, including Ala-substituted peptides, were synthesized. Platelet assays were conducted on healthy donor samples for aggregation, cytotoxicity, adhesion, spreading, and secretion. Mice were used for tail bleeding and FeCl3-induced thrombosis experiments. Clot retraction was assessed using platelet-rich plasma. Extracellular segments of CEACAM1 and A1 domain-derived peptide QDTT were identified, while N, A2, and B domains showed no involvement. QDTT inhibited platelet aggregation. Ala substitution for essential amino acids (Asp139, Thr141, Tyr142, Trp144, and Trp145) in the QDTT sequence abrogated collagen-induced aggregation inhibition. QDTT also suppressed platelet secretion and "inside-out" GP IIb/IIIa activation by convulxin, along with inhibiting PI3K/Akt pathways. QDTT curtailed FeCl3-induced mesenteric thrombosis without significantly prolonging bleeding time, implying the potential of CEACAM1 A1 domain against platelet activation without raising bleeding risk, thus paving the way for novel antiplatelet drugs.
Collapse
Affiliation(s)
- Yujia Ye
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Min Leng
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Shengjie Chai
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Lihong Yang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Longcheng Ren
- Cardiovascular Department, Tengchong Hospital of Traditional Chinese Medicine, Tengchong, PR China
| | - Wen Wan
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Huawei Wang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Longjun Li
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Chaozhong Li
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Zhaohui Meng
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, PR China
| |
Collapse
|
3
|
Oh EB, Shin HJ, Yu H, Jang J, Park JW, Chang TS. NADPH oxidase 1/4 dual inhibitor setanaxib suppresses platelet activation and thrombus formation. Life Sci 2024; 357:123061. [PMID: 39293714 DOI: 10.1016/j.lfs.2024.123061] [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: 07/06/2024] [Revised: 09/07/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
AIMS The production of reactive oxygen species (ROS) by NADPH oxidase (NOX) is able to induce platelet activation, making NOX a promising target for antiplatelet therapy. In this study, we examined the effects of setanaxib, a dual NOX1/4 inhibitor, on human platelet function and ROS-related signaling pathways. MATERIALS AND METHODS In collagen-stimulated human platelets, aggregometry, assessment of ROS and Ca2+, immunoblotting, ELISA, flow cytometry, platelet adhesion assay, and assessment of mouse arterial thrombosis were performed in this study. KEY FINDINGS Setanaxib inhibited both intracellular and extracellular ROS production in collagen-activated platelets. Additionally, setanaxib significantly inhibited collagen-induced platelet aggregation, P-selectin exposure from α-granule release, and ATP release from dense granules. Setanaxib blocked the specific tyrosine phosphorylation-mediated activation of Syk, LAT, Vav1, and Btk within collagen receptor signaling pathways, leading to reduced activation of PLCγ2, PKC, and Ca2+ mobilization. Setanaxib also inhibited collagen-induced activation of integrin αIIbβ3, which is linked to increased cGMP levels and VASP phosphorylation. Furthermore, setanaxib suppressed collagen-induced p38 MAPK activation, resulting in decreased phosphorylation of cytosolic PLA2 and reduced TXA2 generation. Setanaxib also inhibited ERK5 activation, affecting the exposure of procoagulant phosphatidylserine. Setanaxib reduced thrombus formation under shear conditions by preventing platelet adhesion to collagen. Finally, in vivo administration of setanaxib in animal models led to the inhibition of arterial thrombosis. SIGNIFICANCE This study is the first to show that setanaxib suppresses ROS generation, platelet activation, and collagen-induced thrombus formation, suggesting its potential use in treating thrombotic or cardiovascular diseases.
Collapse
Affiliation(s)
- Eun Bee Oh
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| | - Hye Ji Shin
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| | - Hyunseong Yu
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| | - Joara Jang
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| | - Ji Won Park
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| | - Tong-Shin Chang
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
4
|
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
|
5
|
Zhang T, Zhang M, Guo L, Liu D, Zhang K, Bi C, Zhang P, Wang J, Fan Y, He Q, Chang ACY, Zhang J. Angiopoietin-like protein 2 inhibits thrombus formation. Mol Cell Biochem 2024:10.1007/s11010-024-05034-9. [PMID: 38880861 DOI: 10.1007/s11010-024-05034-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/10/2024] [Indexed: 06/18/2024]
Abstract
Acute myocardial infarction is mainly caused by a lack of blood flood in the coronary artery. Angiopoietin-like protein 2 (ANGPTL2) induces platelet activation and thrombus formation in vitro through binding with immunoglobulin-like receptor B, an immunoglobulin superfamily receptor. However, the mechanism by which it regulates platelet function in vivo remains unclear. In this study, we investigated the role of ANGPTL2 during thrombosis in relationship with ST-segment elevation myocardial infarction (STEMI) with spontaneous recanalization (SR). In a cohort of 276 male and female patients, we measured plasma ANGPTL2 protein levels. Using male Angptl2-knockout and wild-type mice, we examined the inhibitory effect of Angptl2 on thrombosis and platelet activation both in vivo and ex vivo. We found that plasma and platelet ANGPTL2 levels were elevated in patients with STEMI with SR compared to those in non-SR (NSR) patients, and was an independent predictor of SR. Angptl2 deficiency accelerated mesenteric artery thrombosis induced by FeCl3 in Angptl2-/- compared to WT animals, promoted platelet granule secretion and aggregation induced by thrombin and collogen while purified ANGPTL2 protein supplementation reversed collagen-induced platelet aggregation. Angptl2 deficiency also increased platelet spreading on immobilized fibrinogen and clot contraction. In collagen-stimulated Angptl2-/- platelets, Src homology region 2 domain-containing phosphatase (Shp)1-Y564 and Shp2-Y580 phosphorylation were attenuated while Src, Syk, and Phospholipase Cγ2 (PLCγ2) phosphorylation increased. Our results demonstrate that ANGPTL2 negatively regulated thrombus formation by activating ITIM which can suppress ITAM signaling pathway. This new knowledge provides a new perspective for designing future antiplatelet aggregation therapies.
Collapse
Affiliation(s)
- Tiantian Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Mingliang Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Lingyu Guo
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Dongsheng Liu
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Kandi Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Changlong Bi
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Peng Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jin Wang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yuqi Fan
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Qing He
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Alex C Y Chang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Junfeng Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| |
Collapse
|
6
|
Nicolai L, Pekayvaz K, Massberg S. Platelets: Orchestrators of immunity in host defense and beyond. Immunity 2024; 57:957-972. [PMID: 38749398 DOI: 10.1016/j.immuni.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 04/06/2024] [Accepted: 04/12/2024] [Indexed: 06/05/2024]
Abstract
Platelets prevent blood loss during vascular injury and contribute to thrombus formation in cardiovascular disease. Beyond these classical roles, platelets are critical for the host immune response. They guard the vasculature against pathogens via specialized receptors, intracellular signaling cascades, and effector functions. Platelets also skew inflammatory responses by instructing innate immune cells, support adaptive immunosurveillance, and influence antibody production and T cell polarization. Concomitantly, platelets contribute to tissue reconstitution and maintain vascular function after inflammatory challenges. However, dysregulated activation of these multitalented cells exacerbates immunopathology with ensuing microvascular clotting, excessive inflammation, and elevated risk of macrovascular thrombosis. This dichotomy underscores the critical importance of precisely defining and potentially modulating platelet function in immunity.
Collapse
Affiliation(s)
- Leo Nicolai
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Kami Pekayvaz
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
7
|
Authi KS, Khan S, Gibbins JM, Brain SD. Evidence that inositol 1,4,5-trisphosphate 3-kinase and inositol 1,3,4,5-tetrakisphosphate are negative regulators of platelet function. Res Pract Thromb Haemost 2024; 8:102326. [PMID: 38404940 PMCID: PMC10885593 DOI: 10.1016/j.rpth.2024.102326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/21/2023] [Accepted: 01/03/2024] [Indexed: 02/27/2024] Open
Abstract
Background Inositol 1,3,4,5-tetrakisphosphate (IP4) is formed from inositol 1,4,5-trisphosphate (IP3) by IP3 3-kinase (ITPK) in most cells. Its function is unknown but has been suggested to be involved in Ca2+ entry, IP3 regulation, and phosphoinositide 3-kinase antagonism. Objectives To better elucidate a function for IP4, we tested a specific inhibitor of ITPK (GNF362) on platelets, the effects of IP4 directly in permeabilized platelets and its effect on phosphatidylinositol 3,4,5-trisphosphate (PIP3) binding to pleckstrin-homology (PH) domain-containing proteins in platelets. Methods Human platelets were utilized in whole blood for thrombus formation, in platelet-rich plasma and washed suspensions for aggregation, and for Ca2+ studies, or resuspended in high K+ and low Na+ buffers for permeabilization experiments. Phosphorylation of AKT-Ser473 and Rap1-GTP formation were measured by Western blotting and PIP3 binding using PIP3 beads. Results GNF362-enhanced platelet aggregation stimulated by low concentrations of ADP, collagen, thrombin, U46619, and thrombus formation in collagen-coated capillaries. GNF362 induced a transient elevation of Ca2+ concentration, elevated basal levels of IP3, and enhanced the peak height of Ca2+ elevated by agonists. In permeabilized platelets, IP4 inhibited GTPγS induced formation of AKT-Ser473 phosphorylation and platelet aggregation. IP4 reduced GTPγS-stimulated Rap1-GTP levels and potently reduced extraction of RASA3 and BTK by PIP3 beads. Conclusion ITPK and IP4 are negative regulators of platelet function. IP4 regulation of PH domain-containing proteins may represent a pathway by which platelet activation may be controlled during thrombosis.
Collapse
Affiliation(s)
- Kalwant S. Authi
- School of Cardiovascular and Metabolic Medicine and Sciences, BHF Centre for Research Excellence, London, UK
| | - Sabeeya Khan
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK
| | - Jonathan M. Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK
| | - Susan D. Brain
- School of Cardiovascular and Metabolic Medicine and Sciences, BHF Centre for Research Excellence, London, UK
| |
Collapse
|
8
|
Li P, Xu B, Xu J, Xu Y, Wang Y, Chen C, Liu P. Lenalidomide Promotes Thrombosis Formation, but Does Not Affect Platelet Activation in Multiple Myeloma. Int J Mol Sci 2023; 24:14097. [PMID: 37762399 PMCID: PMC10532040 DOI: 10.3390/ijms241814097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Lenalidomide, a well-established drug for the treatment of multiple myeloma, significantly enhances patients' survival. Previous clinical studies have demonstrated that its main side effect is an increased risk of thrombotic events. However, the underlying mechanism remains unexplored. Therefore, this study aims to elucidate the mechanism and offer insights into the selection of clinical thrombotic prophylaxis drugs. Firstly, we conducted a retrospective analysis of clinical data from 169 newly diagnosed multiple myeloma patients who received lenalidomide. To confirm the impact of lenalidomide on thrombosis formation, FeCl3-induced thrombosis and deep venous thrombosis models in mice were established. To investigate the effects of lenalidomide on platelet function, both in vivo and in vitro experiments were designed. During the follow-up period, 8 patients developed thrombotic events, including 8 venous and 1 arterial. Further investigation using mice models demonstrated that lenalidomide significantly promoted the formation of venous thrombosis, consistent with clinical findings. To elucidate the underlying mechanism, assays were conducted to assess platelet function and coagulation. We observed that lenalidomide did not have any noticeable impact on platelet function, both in vitro and in vivo, while administration of lenalidomide resulted in significant decreases in prothrombin time, thrombin time, and prothrombin time ratio in patients, as well as a remarkable reduction in tail-bleeding time in mice. The administration of lenalidomide had no significant impact on platelet function, which may affect venous thrombus formation by affecting coagulation. Therefore, anticoagulant drugs may be superior to antiplatelet drugs in the selection of clinical thrombus prophylaxis.
Collapse
Affiliation(s)
- Panpan Li
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (P.L.); (B.X.); (J.X.); (Y.W.); (C.C.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Bei Xu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (P.L.); (B.X.); (J.X.); (Y.W.); (C.C.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiadai Xu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (P.L.); (B.X.); (J.X.); (Y.W.); (C.C.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yanyan Xu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200032, China;
| | - Yawen Wang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (P.L.); (B.X.); (J.X.); (Y.W.); (C.C.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chen Chen
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (P.L.); (B.X.); (J.X.); (Y.W.); (C.C.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Peng Liu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (P.L.); (B.X.); (J.X.); (Y.W.); (C.C.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
9
|
Kukaliia ON, Ageev SV, Petrov AV, Kirik OV, Korzhevskii DE, Meshcheriakov AA, Jakovleva AA, Poliakova LS, Novikova TA, Kolpakova ME, Vlasov TD, Molchanov OE, Maistrenko DN, Murin IV, Sharoyko VV, Semenov KN. C 60 adduct with L-arginine as a promising nanomaterial for treating cerebral ischemic stroke. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 53:102698. [PMID: 37507062 DOI: 10.1016/j.nano.2023.102698] [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: 03/24/2023] [Revised: 07/03/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023]
Abstract
The work aimed to investigate the biocompatibility and biological activity of the water-soluble fullerene adduct C60-Arg. It was found that the material is haemocompatible, is not cyto- and genotoxic, possesses pronounced antioxidant activity. Additionally, this paper outlines the direction of application of water-soluble fullerene adducts in the creation of neuroprotectors. It has been suggested that a putative mechanism of the protective action of the C60-Arg adduct is associated with its antioxidant properties, the ability to penetrate the blood-brain barrier, and release nitrogen monoxide as a result of the catabolism of L-arginine residues, which promote vascular relaxation. The action of the C60-Arg adduct was compared with the action of such an antioxidant as Edaravone, which is approved in Japan for the treatment of ischemic and haemorrhagic strokes.
Collapse
Affiliation(s)
- Olegi N Kukaliia
- Pavlov First Saint Petersburg State Medical University, 6-8 L'va Tolstogo Str., Saint Petersburg, 197022, Russia
| | - Sergei V Ageev
- Pavlov First Saint Petersburg State Medical University, 6-8 L'va Tolstogo Str., Saint Petersburg, 197022, Russia; Institute of Chemistry, Saint Petersburg State University, 26 Universitetskii Pr., Saint Petersburg, 198504, Russia
| | - Andrey V Petrov
- Institute of Chemistry, Saint Petersburg State University, 26 Universitetskii Pr., Saint Petersburg, 198504, Russia
| | - Olga V Kirik
- Institute of Experimental Medicine, 12 Akademika Pavlova Str., Saint Petersburg, 197022, Russia
| | - Dmitrii E Korzhevskii
- Institute of Experimental Medicine, 12 Akademika Pavlova Str., Saint Petersburg, 197022, Russia
| | - Anatolii A Meshcheriakov
- Pavlov First Saint Petersburg State Medical University, 6-8 L'va Tolstogo Str., Saint Petersburg, 197022, Russia
| | - Anastasia A Jakovleva
- Pavlov First Saint Petersburg State Medical University, 6-8 L'va Tolstogo Str., Saint Petersburg, 197022, Russia
| | - Liudmila S Poliakova
- Pavlov First Saint Petersburg State Medical University, 6-8 L'va Tolstogo Str., Saint Petersburg, 197022, Russia
| | - Tatiana A Novikova
- Pavlov First Saint Petersburg State Medical University, 6-8 L'va Tolstogo Str., Saint Petersburg, 197022, Russia
| | - Maria E Kolpakova
- Pavlov First Saint Petersburg State Medical University, 6-8 L'va Tolstogo Str., Saint Petersburg, 197022, Russia
| | - Timur D Vlasov
- Pavlov First Saint Petersburg State Medical University, 6-8 L'va Tolstogo Str., Saint Petersburg, 197022, Russia
| | - Oleg E Molchanov
- A. M. Granov Russian Research Centre for Radiology and Surgical Technologies, 70 Leningradskaia Str., Saint Petersburg, 197758, Russia
| | - Dmitriy N Maistrenko
- A. M. Granov Russian Research Centre for Radiology and Surgical Technologies, 70 Leningradskaia Str., Saint Petersburg, 197758, Russia
| | - Igor V Murin
- Institute of Chemistry, Saint Petersburg State University, 26 Universitetskii Pr., Saint Petersburg, 198504, Russia
| | - Vladimir V Sharoyko
- Pavlov First Saint Petersburg State Medical University, 6-8 L'va Tolstogo Str., Saint Petersburg, 197022, Russia; Institute of Chemistry, Saint Petersburg State University, 26 Universitetskii Pr., Saint Petersburg, 198504, Russia; A. M. Granov Russian Research Centre for Radiology and Surgical Technologies, 70 Leningradskaia Str., Saint Petersburg, 197758, Russia.
| | - Konstantin N Semenov
- Pavlov First Saint Petersburg State Medical University, 6-8 L'va Tolstogo Str., Saint Petersburg, 197022, Russia; Institute of Chemistry, Saint Petersburg State University, 26 Universitetskii Pr., Saint Petersburg, 198504, Russia; A. M. Granov Russian Research Centre for Radiology and Surgical Technologies, 70 Leningradskaia Str., Saint Petersburg, 197758, Russia.
| |
Collapse
|
10
|
Wang L, Liu Y, Tian R, Zuo W, Qian H, Wang L, Yang X, Liu Z, Zhang S. What do we know about platelets in myocardial ischemia-reperfusion injury and why is it important? Thromb Res 2023; 229:114-126. [PMID: 37437517 DOI: 10.1016/j.thromres.2023.06.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/22/2023] [Accepted: 06/23/2023] [Indexed: 07/14/2023]
Abstract
Myocardial ischemia-reperfusion injury (MIRI), the joint result of ischemic injury and reperfusion injury, is associated with poor outcomes in patients with acute myocardial infarction undergoing primary percutaneous coronary intervention. Accumulating evidence demonstrates that activated platelets directly contribute to the pathogenesis of MIRI through participating in the formation of microthrombi, interaction with leukocytes, secretion of active substances, constriction of microvasculature, and activation of spinal afferent nerves. The molecular mechanisms underlying the above detrimental effects of activated platelets include the homotypic and heterotypic interactions through surface receptors, transduction of intracellular signals, and secretion of active substances. Revealing the roles of platelet activation in MIRI and the associated mechanisms would provide potential targets/strategies for the clinical evaluation and treatment of MIRI. Further studies are needed to characterize the temporal (ischemia phase vs. reperfusion phase) and spatial (systemic vs. local) distributions of platelet activation in MIRI by multi-omics strategies. To improve the likelihood of translating novel cardioprotective interventions into clinical practice, basic researches maximally replicating the complexity of clinical scenarios would be necessary.
Collapse
Affiliation(s)
- Lun Wang
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Yifan Liu
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Ran Tian
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Wei Zuo
- Department of Pharmacy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Hao Qian
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Liang Wang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Xinglin Yang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Zhenyu Liu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China.
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
11
|
Janus-Bell E, Mangin PH. The relative importance of platelet integrins in hemostasis, thrombosis and beyond. Haematologica 2023; 108:1734-1747. [PMID: 36700400 PMCID: PMC10316258 DOI: 10.3324/haematol.2022.282136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Integrins are heterodimeric transmembrane receptors composed of α and β chains, with an N-terminal extracellular domain forming a globular head corresponding to the ligand binding site. Integrins regulate various cellular functions including adhesion, migration, proliferation, spreading and apoptosis. On platelets, integrins play a central role in adhesion and aggregation on subendothelial matrix proteins of the vascular wall, thereby ensuring hemostasis. Platelet integrins belong either to the β1 family (α2β1, α5β1 and α6β1) or to the β3 family (αIIbβ3 and αvβ3). On resting platelets, integrins can engage their ligands when the latter are immobilized but not in their soluble form. The effects of various agonists promote an inside-out signal in platelets, increasing the affinity of integrins for their ligands and conveying a modest signal reinforcing platelet activation, called outside-in signaling. This outside-in signal ensures platelet adhesion, shape change, granule secretion and aggregation. In this review, we examine the role of each platelet integrin in hemostatic plug formation, hemostasis and arterial thrombosis and also beyond these classical functions, notably in tumor metastasis and sepsis.
Collapse
Affiliation(s)
- Emily Janus-Bell
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, F-67065 Strasbourg.
| | - Pierre H Mangin
- Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S1255, FMTS, F-67065 Strasbourg
| |
Collapse
|
12
|
Cheung HYF, Zou J, Tantiwong C, Fernandez DI, Huang J, Ahrends R, Roest M, Cavill R, Gibbins J, Heemskerk JWM. High-throughput assessment identifying major platelet Ca 2+ entry pathways via tyrosine kinase-linked and G protein-coupled receptors. Cell Calcium 2023; 112:102738. [PMID: 37060673 DOI: 10.1016/j.ceca.2023.102738] [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: 12/01/2022] [Revised: 03/04/2023] [Accepted: 04/06/2023] [Indexed: 04/17/2023]
Abstract
In platelets, elevated cytosolic Ca2+ is a crucial second messenger, involved in most functional responses, including shape change, secretion, aggregation and procoagulant activity. The platelet Ca2+ response consists of Ca2+ mobilization from endoplasmic reticulum stores, complemented with store-operated or receptor-operated Ca2+ entry pathways. Several channels can contribute to the Ca2+ entry, but their relative contribution is unclear upon stimulation of ITAM-linked receptors such as glycoprotein VI (GPVI) and G-protein coupled receptors such as the protease-activated receptors (PAR) for thrombin. We employed a 96-well plate high-throughput assay with Fura-2-loaded human platelets to perform parallel [Ca2+]i measurements in the presence of EGTA or CaCl2. Per agonist condition, this resulted in sets of EGTA, CaCl2 and Ca2+ entry ratio curves, defined by six parameters, reflecting different Ca2+ ion fluxes. We report that threshold stimulation of GPVI or PAR, with a variable contribution of secondary mediators, induces a maximal Ca2+ entry ratio of 3-7. Strikingly, in combination with Ca2+-ATPase inhibition by thapsigargin, the maximal Ca2+ entry ratio increased to 400 (GPVI) or 40 (PAR), pointing to a strong receptor-dependent enhancement of store-operated Ca2+ entry. By pharmacological blockage of specific Ca2+ channels in platelets, we found that, regardless of GPVI or PAR stimulation, the Ca2+ entry ratio was strongest affected by inhibition of ORAI1 (2-APB, Synta66) > Na+/Ca2+ exchange (NCE) > P2×1 (only initial). In contrast, inhibition of TRPC6, Piezo1/2 or STIM1 was without effect. Together, these data reveal ORAI1 and NCE as dominating Ca2+ carriers regulating GPVI- and PAR-induced Ca2+ entry in human platelets.
Collapse
Affiliation(s)
- Hilaire Yam Fung Cheung
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands; Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V, Dortmund, Germany; Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jinmi Zou
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands; Synapse Research Institute Maastricht, 6217 KD Maastricht, The Netherlands
| | - Chukiat Tantiwong
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands; Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Delia I Fernandez
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands; Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, 15706, Spain
| | - Jingnan Huang
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands; Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V, Dortmund, Germany; Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, 15706, Spain
| | - Robert Ahrends
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V, Dortmund, Germany; Dept. of Analytical Chemistry, University of Vienna, Vienna, Austria
| | - Mark Roest
- Synapse Research Institute Maastricht, 6217 KD Maastricht, The Netherlands
| | - Rachel Cavill
- Department of Advanced Computing Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Jon Gibbins
- Institute for Cardiovascular and Metabolic Research (ICMR), School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands; Synapse Research Institute Maastricht, 6217 KD Maastricht, The Netherlands.
| |
Collapse
|
13
|
Li L, Xu X, Lv K, Zheng G, Wang H, Chen S, Huang L, Liu Y, Zhang Y, Tang Z, Zhang L, Wang J, Qiao J, Li H, Wang X, Yao G, Fang C. Asebogenin suppresses thrombus formation via inhibition of Syk phosphorylation. Br J Pharmacol 2023; 180:287-307. [PMID: 36166754 DOI: 10.1111/bph.15964] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/24/2022] [Accepted: 09/11/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Thrombosis is a major cause of morbidity and mortality worldwide. Platelet activation by exposed collagen through glycoprotein VI (GPVI) and formation of neutrophil extracellular traps (NETs) are critical pathogenic factors for arterial and venous thrombosis. Both events are regulated by spleen tyrosine kinase (Syk)-mediated signalling events. Asebogenin is a dihydrochalcone whose pharmacological effects remain largely unknown. This study aims to investigate the antithrombotic effects of asebogenin and the underlying molecular mechanisms. EXPERIMENTAL APPROACH Platelet aggregation was assessed using an aggregometer. Platelet P-selectin exposure, integrin activation and calcium mobilization were determined by flow cytometry. NETs formation was assessed by SYTOX Green staining and immunohistochemistry. Quantitative phosphoproteomics, microscale thermophoresis, in vitro kinase assay and molecular docking combined with dynamics simulation were performed to characterize the targets of asebogenin. The in vivo effects of asebogenin on arterial thrombosis were investigated using FeCl3 -induced and laser-induced injury models, whereas those of venous thrombosis were induced by stenosis of the inferior vena cava. KEY RESULTS Asebogenin inhibited a series of GPVI-induced platelet responses and suppressed NETs formation induced by proinflammatory stimuli. Mechanistically, asebogenin directly interfered with the phosphorylation of Syk at Tyr525/526, which is important for its activation. Further, asebogenin suppressed arterial thrombosis demonstrated by decreased platelet accumulation and fibrin generation and attenuated venous thrombosis determined by reduced neutrophil accumulation and NETs formation, without increasing bleeding risk. CONCLUSION AND IMPLICATIONS Asebogenin exhibits potent antithrombotic effects by targeting Syk and is a potential lead compound for the development of efficient and safe antithrombotic agents.
Collapse
Affiliation(s)
- Li Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xulin Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Keyu Lv
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guijuan Zheng
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hao Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuai Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lang Huang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Liu
- DeepKinase Biotechnologies Ltd., Beijing, China
| | | | - Zhaoming Tang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lili Zhang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jinyu Wang
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,The Key Laboratory of Oral and Maxillofacial Development and Regeneration of Hubei Province, Wuhan, Hubei, China
| | - Jianlin Qiao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongliang Li
- Laboratory of Chinese Herbal Pharmacology, Department of Pharmacy, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China.,Biomedical Research Institute, School of Pharmaceutical Sciences and Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xuanbin Wang
- Laboratory of Chinese Herbal Pharmacology, Department of Pharmacy, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China.,Biomedical Research Institute, School of Pharmaceutical Sciences and Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Guangmin Yao
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chao Fang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei, China
| |
Collapse
|
14
|
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
|
15
|
An optimized herbal medicine containing Scutellaria baicalensis Georgi, Alisma orientale Juzepzuk, and Atractylodes japonica Koidzumi has potent antiplatelet and antithrombotic activities. J Tradit Complement Med 2023; 13:285-296. [PMID: 37128192 PMCID: PMC10148138 DOI: 10.1016/j.jtcme.2023.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/11/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Background and aim Platelet-derived thrombosis is important in the pathogenesis of cardiovascular diseases. HTB is an optimized herbal medicine including Scutellaria baicalensis Georgi, Alisma orientale Juzepzuk, and Atractylodes japonica Koidzumi. It is widely used in traditional medicine due to its anti-inflammatory and antioxidant effects. However, its antiplatelet and antithrombotic activities have not been completely validated. The current study aimed to examine the inhibitory effect of the novel herb formula HTB against platelet activation and thrombus formation. Experimental procedure The antiplatelet activities of HTB via platelet aggregation, granule secretion, reactive oxygen species generation, and intracellular calcium mobilization were evaluated. Moreover, the antithrombotic effect of HTB via FeCl3-induced arterial thrombus formation in vivo in mice was assessed. The inhibitory effect of HTB against primary hemostasis was investigated based on transection tail bleeding time. Results and conclusion HTB treatment significantly inhibited glycoprotein VI-mediated platelet aggregation, granule secretion, reactive oxygen species generation, and intracellular calcium mobilization. Biochemical studies revealed that HTB inhibited glycoprotein VI-mediated platelet signal transduction during cell activation. Further, its antioxidant effect might be derived by reducing the phosphorylation of the p47phox/Hic5 axis signalosome. Oral HTB treatment was effective in decreasing FeCl3-induced arterial thrombus formation without prolonging the tail bleeding time. HTB can be an effective therapeutic agent against thrombotic diseases.
Collapse
|
16
|
How snake venom disintegrins affect platelet aggregation and cancer proliferation. Toxicon 2022; 221:106982. [DOI: 10.1016/j.toxicon.2022.106982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022]
|
17
|
Zheng TJ, Parra-Izquierdo I, Reitsma SE, Heinrich MC, Larson MK, Shatzel JJ, Aslan JE, McCarty OJT. Platelets and tyrosine kinase inhibitors: clinical features, mechanisms of action, and effects on physiology. Am J Physiol Cell Physiol 2022; 323:C1231-C1250. [PMID: 35938677 PMCID: PMC9576167 DOI: 10.1152/ajpcell.00040.2022] [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: 02/01/2022] [Revised: 08/01/2022] [Accepted: 08/01/2022] [Indexed: 11/22/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) have emerged as a promising class of target-directed, small molecule inhibitors used to treat hematologic malignancies, inflammatory diseases, and autoimmune disorders. Recently, TKIs have also gained interest as potential antiplatelet-directed therapeutics that could be leveraged to reduce pathologic thrombus formation and atherothrombotic complications, while minimally affecting platelet hemostatic function. This review provides a mechanistic overview and summarizes the known effects of tyrosine kinase inhibitors on platelet signaling and function, detailing prominent platelet signaling pathways downstream of the glycoprotein VI (GPVI) receptor, integrin αIIbβ3, and G protein-coupled receptors (GPCRs). This review focuses on mechanistic as well as clinically relevant and emerging TKIs targeting major families of tyrosine kinases including but not limited to Bruton's tyrosine kinase (BTK), spleen tyrosine kinase (Syk), Src family kinases (SFKs), Janus kinases (JAK), and signal transducers and activators of transcription (STAT) and evaluates their effects on platelet aggregation and adhesion, granule secretion, receptor expression and activation, and protein phosphorylation events. In summation, this review highlights current advances and knowledge on the effects of select TKIs on platelet biology and furthers insight on signaling pathways that may represent novel druggable targets coupled to specific platelet functional responses.
Collapse
Affiliation(s)
- Tony J Zheng
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Iván Parra-Izquierdo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Stéphanie E Reitsma
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Michael C Heinrich
- Portland Veterans Affairs Health Care System and Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Department of Molecular and Cellular Biosciences, Oregon Health & Science University, Portland, Oregon
| | - Mark K Larson
- Department of Biology, Augustana University, Sioux Falls, South Dakota
| | - Joseph J Shatzel
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
- Division of Hematology & Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Joseph E Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
- Division of Hematology & Medical Oncology, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental & Cancer Biology, School of Medicine, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
18
|
Protein tyrosine phosphatase PTPN22 negatively modulates platelet function and thrombus formation. Blood 2022; 140:1038-1051. [PMID: 35767715 DOI: 10.1182/blood.2022015554] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/21/2022] [Indexed: 11/20/2022] Open
Abstract
Protein tyrosine phosphatase nonreceptor type 22 (PTPN22) is a protein tyrosine phosphatase that negatively regulates T-cell signaling. However, whether it is expressed and functions in platelets remains unknown. Here we investigated the expression and role of PTPN22 in platelet function. We reported PTPN22 expression in both human and mouse platelets. Using PTPN22-/- mice, we showed that PTPN22 deficiency significantly shortened tail-bleeding time and accelerated arterial thrombus formation without affecting venous thrombosis and the coagulation factors VIII and IX. Consistently, PTPN22-deficient platelets exhibited enhanced platelet aggregation, granule secretion, calcium mobilization, lamellipodia formation, spreading, and clot retraction. Quantitative phosphoproteomic analysis revealed the significant difference of phosphodiesterase 5A (PDE5A) phosphorylation in PTPN22-deficient platelets compared with wild-type platelets after collagen-related peptide stimulation, which was confirmed by increased PDE5A phosphorylation (Ser92) in collagen-related peptide-treated PTPN22-deficient platelets, concomitant with reduced level and vasodilator-stimulated phosphoprotein phosphorylation (Ser157/239). In addition, PTPN22 interacted with phosphorylated PDE5A (Ser92) and dephosphorylated it in activated platelets. Moreover, purified PTPN22 but not the mutant form (C227S) possesses intrinsic serine phosphatase activity. Furthermore, inhibition of PTPN22 enhanced human platelet aggregation, spreading, clot retraction, and increased PDE5A phosphorylation (Ser92). In conclusion, our study shows a novel role of PTPN22 in platelet function and arterial thrombosis, identifying new potential targets for future prevention of thrombotic or cardiovascular diseases.
Collapse
|
19
|
Oxidised Low-Density Lipoprotein-Induced Platelet Hyperactivity—Receptors and Signalling Mechanisms. Int J Mol Sci 2022; 23:ijms23169199. [PMID: 36012465 PMCID: PMC9409144 DOI: 10.3390/ijms23169199] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/26/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
Dyslipidaemia leads to proatherogenic oxidative lipid stress that promotes vascular inflammation and thrombosis, the pathologies that underpin myocardial infarction, stroke, and deep vein thrombosis. These prothrombotic states are driven, at least in part, by platelet hyperactivity, and they are concurrent with the appearancxe of oxidatively modified low-density lipoproteins (LDL) in the circulation. Modified LDL are heterogenous in nature but, in a general sense, constitute a prototype circulating transporter for a plethora of oxidised lipid epitopes that act as danger-associated molecular patterns. It is well-established that oxidatively modified LDL promote platelet activation and arterial thrombosis through a number of constitutively expressed scavenger receptors, which transduce atherogenic lipid stress to a complex array of proactivatory signalling pathways in the platelets. Stimulation of these signalling events underlie the ability of modified LDL to induce platelet activation and blunt platelet inhibitory pathways, as well as promote platelet-mediated coagulation. Accumulating evidence from patients at risk of arterial thrombosis and experimental animal models of disease suggest that oxidised LDL represents a tangible link between the dyslipidaemic environment and increased platelet activation. The aim of this review is to summarise recent advances in our understanding of the pro-thrombotic signalling events induced in platelets by modified LDL ligation, describe the contribution of individual platelet scavenger receptors, and highlight potential future challenges of targeting these pathways.
Collapse
|
20
|
Kostyak JC, Mauri B, Dangelmaier C, Vari HR, Patel A, Wright M, Reddy H, Tsygankov AY, Kunapuli SP. Phosphorylation on Syk Y342 is important for both ITAM and hemITAM signaling in platelets. J Biol Chem 2022; 298:102189. [PMID: 35753354 PMCID: PMC9287148 DOI: 10.1016/j.jbc.2022.102189] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 11/29/2022] Open
Abstract
Immune cells express receptors bearing an immune tyrosine activation motif (ITAM) containing two YXXL motifs or hemITAMs containing only one YXXL motif. Phosphorylation of the ITAM/hemITAM is mediated by Src family kinases allowing for the binding and activation of spleen tyrosine kinase (Syk). It is believed that Syk must be phosphorylated on tyrosine residues for activation, and Tyr342, а conserved tyrosine in the interdomain B region, has been shown to be critical for regulating Syk in FcεR1-activated mast cells. Syk is a key mediator of signaling pathways downstream of several platelet pathways including the ITAM bearing glycoprotein VI (GPVI)/Fc receptor gamma chain collagen receptor and the hemITAM containing C-type lectin-like receptor-2 (CLEC-2). Since platelet activation is a crucial step in both hemostasis and thrombosis, we evaluated the importance of Syk Y342 in these processes by producing an Syk Y342F knock-in mouse. When using a CLEC-2 antibody as an agonist, reduced aggregation and secretion were observed in Syk Y342F mouse platelets when compared with control mouse platelets. Platelet reactivity was also reduced in response to the GPVI agonist collagen-related peptide. Signaling initiated by either GPVI or CLEC-2 was also greatly inhibited, including Syk Y519/520 phosphorylation. Hemostasis, as measured by tail bleeding time, was not altered in Syk Y342F mice, but thrombus formation in response to FeCl3 injury was prolonged in Syk Y342F mice. These data demonstrate that phosphorylation of Y342 on Syk following stimulation of either GPVI or CLEC-2 receptors is important for the ability of Syk to transduce a signal.
Collapse
Affiliation(s)
- John C Kostyak
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Benjamin Mauri
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Carol Dangelmaier
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Hymavathi Reddy Vari
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Akruti Patel
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Monica Wright
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Haritha Reddy
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Alexander Y Tsygankov
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Satya P Kunapuli
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
21
|
Lee MJ, Weng CM, Chao W, Fang YF, Chung FT, Lin CH, Kuo HP. Platelet Activation in High D-Dimer Plasma Plays a Role in Acquired Resistance to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Patients with Mutant Lung Adenocarcinoma. Front Oncol 2022; 12:876051. [PMID: 35756605 PMCID: PMC9214222 DOI: 10.3389/fonc.2022.876051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/09/2022] [Indexed: 11/21/2022] Open
Abstract
Objective Platelet activation and adhesion to cancer cells increase the release of multiple factors that contribute to EMT and chemoresistance. Elevated levels of D-dimer have been associated with poor clinical outcomes in lung cancer. Platelets in high D-dimer plasma may be activated and implicated in acquired resistance to EGFR TKI in advanced lung adenocarcinoma with mutant EGFR. Materials and Methods Clinical responsive rate (RR), progression-free survival (PFS), and overall survival (OS) were prospectively measured in treatment-naïve lung adenocarcinoma patients with activation mutation. Plasma or platelets from patients with high or low D-dimer level were obtained to investigate the cytotoxic effects of TKIs on mutant cancer cells, and the mechanistic pathways were also explored. Results Patients with high D-dimer had worse RR, PFS, and OS. High D-dimer plasma induced resistance to gefitinib, erlotinib, afatinib, or osimertinib in EGFR mutant lung cancer cells. Depletion of platelets in high D-dimer plasma reversed the resistance to TKI. Platelets of high D-dimer plasma had higher adherence capacity to cancer cells, and induced EGFR and Akt activation as well as EMT through Src activation. Inhibition of platelet adherence or activation of Src or Akt conquered the resistance to TKI. The acquired resistance to TKI by high D-dimer plasma was less attributed to secondary gene mutation. Conclusion Increased platelet activation in the high D-dimer plasma may contribute to first-line acquired EGFR TKI resistance. Thus, therapeutic strategy against platelet activation in patients with high D-dimer levels may improve the efficacy of first-line treatment with EGFR TKI.
Collapse
Affiliation(s)
- Meng-Jung Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Thoracic Medicine Research Center, Taipei Medical University, Taipei, Taiwan
| | - Chih-Ming Weng
- Thoracic Medicine Research Center, Taipei Medical University, Taipei, Taiwan.,School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei Chao
- Thoracic Medicine Research Center, Taipei Medical University, Taipei, Taiwan
| | - Yueh-Fu Fang
- Department of Thoracic Medicine, Chang Gung Medical Foundation, Chang Gung University College of Medicine, Taipei, Taiwan
| | - Fu-Tsai Chung
- Department of Thoracic Medicine, Chang Gung Medical Foundation, Chang Gung University College of Medicine, Taipei, Taiwan
| | - Chien-Huang Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Thoracic Medicine Research Center, Taipei Medical University, Taipei, Taiwan
| | - Han-Pin Kuo
- Thoracic Medicine Research Center, Taipei Medical University, Taipei, Taiwan.,Department of Thoracic Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
22
|
Li Y, Xin G, Li S, Dong Y, Zhu Y, Yu X, Wan C, Li F, Wei Z, Wang Y, Zhang K, Chen Q, Niu H, Huang W. PD-L1 Regulates Platelet Activation and Thrombosis via Caspase-3/GSDME Pathway. Front Pharmacol 2022; 13:921414. [PMID: 35784685 PMCID: PMC9240427 DOI: 10.3389/fphar.2022.921414] [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: 04/15/2022] [Accepted: 05/25/2022] [Indexed: 11/24/2022] Open
Abstract
Platelets play a central role in hemostasis and thrombosis, regulating the occurrence and development of thrombotic diseases, including ischemic stroke. Programmed death ligand 1 (PD-L1) has recently been detected in platelet, while the function of PD-L1 in platelets remain elusive. Our data reveal a novel mechanism for the role of PD-L1 on platelet activation and arterial thrombosis. PD-L1 knockout does not affect platelet morphology, count, and mean volume under homeostasis and without risk of bleeding, which inhibits platelet activation by suppressing outside-in-activation of integrin by downregulating the Caspase-3/GSDME pathway. Platelet adoptive transfer experiments demonstrate that PD-L1 knockout inhibits thrombosis. And the absence of PD-L1 improves ischemic stroke severity and increases mice survival. Immunohistochemical staining of the internal structure of the thrombus proves that PD-L1 enhances the seriousness of the thrombus by inhibiting platelet activation. This work reveals a regulatory role of PD-L1 on platelet activation and thrombosis while providing novel platelet intervention strategies to prevent thrombosis.
Collapse
|
23
|
Abbas MN, Chlastáková A, Jmel MA, Iliaki-Giannakoudaki E, Chmelař J, Kotsyfakis M. Serpins in Tick Physiology and Tick-Host Interaction. Front Cell Infect Microbiol 2022; 12:892770. [PMID: 35711658 PMCID: PMC9195624 DOI: 10.3389/fcimb.2022.892770] [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: 03/09/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
Tick saliva has been extensively studied in the context of tick-host interactions because it is involved in host homeostasis modulation and microbial pathogen transmission to the host. Accumulated knowledge about the tick saliva composition at the molecular level has revealed that serine protease inhibitors play a key role in the tick-host interaction. Serpins are one highly expressed group of protease inhibitors in tick salivary glands, their expression can be induced during tick blood-feeding, and they have many biological functions at the tick-host interface. Indeed, tick serpins have an important role in inhibiting host hemostatic processes and in the modulation of the innate and adaptive immune responses of their vertebrate hosts. Tick serpins have also been studied as potential candidates for therapeutic use and vaccine development. In this review, we critically summarize the current state of knowledge about the biological role of tick serpins in shaping tick-host interactions with emphasis on the mechanisms by which they modulate host immunity. Their potential use in drug and vaccine development is also discussed.
Collapse
Affiliation(s)
- Muhammad Nadeem Abbas
- State Key Laboratory of Silkworm Genome Biology, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture, Southwest University, Chongqing, China
| | - Adéla Chlastáková
- Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, České Budějovice, Czechia
- Laboratory of Molecular Biology of Ticks, Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czechia
| | - Mohamed Amine Jmel
- Laboratory of Genomics and Proteomics of Disease Vectors, Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czechia
| | | | - Jindřich Chmelař
- Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, České Budějovice, Czechia
- *Correspondence: Jindřich Chmelař, ; Michail Kotsyfakis,
| | - Michail Kotsyfakis
- Laboratory of Genomics and Proteomics of Disease Vectors, Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czechia
- *Correspondence: Jindřich Chmelař, ; Michail Kotsyfakis,
| |
Collapse
|
24
|
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: 19] [Impact Index Per Article: 6.3] [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
|
25
|
Li L, Roest M, Meijers JCM, de Laat B, Urbanus RT, de Groot PG, Huskens D. Platelet Activation via Glycoprotein VI Initiates Thrombin Generation: A Potential Role for Platelet-Derived Factor IX? Thromb Haemost 2022; 122:1502-1512. [PMID: 35512832 DOI: 10.1055/s-0042-1744379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Collagen triggers coagulation via activation of factor (F) XII. In a platelet-rich environment, collagen can also trigger coagulation independently of FXII. We studied a novel mechanism of coagulation initiation via collagen-dependent platelet activation using thrombin generation (TG) in platelet-rich plasma. Collagen-induced coagulation is minimally affected by active-site inactivated FVIIa, anti-FVII antibodies, or FXIIa inhibition (corn trypsin inhibitor). Activation of platelets via specific glycoprotein (GP) VI agonists initiates TG, FX activation, and fibrin formation. To determine the platelet-derived trigger of coagulation, we systematically reconstituted factor-deficient plasmas with washed platelets. TG triggered by GPVI-activated platelets was significantly affected in FIX- and FVIII-deficient plasma but not in FVII- and FXII-deficient plasma. In a purified system composed of FX and FVIII, we observed that absence of FIX was compensated by GPVI-activated platelets, which could be inhibited by an anti-FIX antibody, suggesting FIXa activity from activated platelets. Furthermore, with the addition of FVIII in FIX-deficient plasma, TG induced by GPVI-activated platelets was restored, and was inhibited by the anti-FIX antibody. In conclusion, GPVI-activated platelets initiate TG, probably via platelet-derived FIXa activity.
Collapse
Affiliation(s)
- Li Li
- Department of Platelet Pathophysiology, Synapse Research Institute, Maastricht, the Netherlands.,Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Mark Roest
- Department of Platelet Pathophysiology, Synapse Research Institute, Maastricht, the Netherlands.,Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Joost C M Meijers
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands.,Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Bas de Laat
- Department of Platelet Pathophysiology, Synapse Research Institute, Maastricht, the Netherlands.,Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Rolf T Urbanus
- Van Creveldkliniek, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Philip G de Groot
- Department of Platelet Pathophysiology, Synapse Research Institute, Maastricht, the Netherlands
| | - Dana Huskens
- Department of Platelet Pathophysiology, Synapse Research Institute, Maastricht, the Netherlands.,Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
26
|
Dangelmaier C, Vari HR, Wright M, Kostyak JC, Kunapuli SP. Clustering extent-dependent differential signaling by CLEC-2 receptors in platelets. Res Pract Thromb Haemost 2022; 6:e12710. [PMID: 35573643 PMCID: PMC9074038 DOI: 10.1002/rth2.12710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/08/2022] [Indexed: 11/12/2022] Open
Abstract
Background C-type lectin receptor family members play a role in many cells including platelets, where they are crucial in the separation of lymphatic and blood vessels during development. The C-type lectin-like receptor 2 (CLEC-2) receptor contains the canonical intracellular hemITAM motif through which it signals to activate Syk. Objectives One proposed hypothesis for signaling cascade is that Syk bridges two receptors through phosphorylated hemITAM motifs. We demonstrated that the phosphorylated hemITAM stimulates PI3 kinase/Btk pathways to activate Syk. To address this controversy, we used a CLEC-2 selective agonist and studied the role of Btk in platelet activation. Results and Conclusions Platelet activation and downstream signaling were abolished in murine and human platelets in the presence of the Btk inhibitors ibrutinib or acalabrutinib when a low concentration of a CLEC-2 antibody was used to crosslink CLEC-2 receptors. This inhibition was overcome by increasing concentrations of the CLEC-2 antibody. Similar results were obtained in X-linked immunodeficient mouse platelets, with an inactivating mutation in Btk or in Lyn null platelets. We conclude that at low crosslinking conditions of CLEC-2, Btk plays an important role in the activation of Syk, but at higher crosslinking conditions their role becomes less important and other mechanisms take over to activate Syk.
Collapse
Affiliation(s)
- Carol Dangelmaier
- Sol Sherry Thrombosis Research Center Lewis Katz School of Medicine Temple University Philadelphia Pennsylvania USA
| | - Hymavathi Reddy Vari
- Sol Sherry Thrombosis Research Center Lewis Katz School of Medicine Temple University Philadelphia Pennsylvania USA
| | - Monica Wright
- Sol Sherry Thrombosis Research Center Lewis Katz School of Medicine Temple University Philadelphia Pennsylvania USA
| | - John C Kostyak
- Sol Sherry Thrombosis Research Center Lewis Katz School of Medicine Temple University Philadelphia Pennsylvania USA
| | - Satya P Kunapuli
- Sol Sherry Thrombosis Research Center Lewis Katz School of Medicine Temple University Philadelphia Pennsylvania USA
| |
Collapse
|
27
|
Veuthey L, Aliotta A, Bertaggia Calderara D, Pereira Portela C, Alberio L. Mechanisms Underlying Dichotomous Procoagulant COAT Platelet Generation-A Conceptual Review Summarizing Current Knowledge. Int J Mol Sci 2022; 23:2536. [PMID: 35269679 PMCID: PMC8910683 DOI: 10.3390/ijms23052536] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 12/23/2022] Open
Abstract
Procoagulant platelets are a subtype of activated platelets that sustains thrombin generation in order to consolidate the clot and stop bleeding. This aspect of platelet activation is gaining more and more recognition and interest. In fact, next to aggregating platelets, procoagulant platelets are key regulators of thrombus formation. Imbalance of both subpopulations can lead to undesired thrombotic or bleeding events. COAT platelets derive from a common pro-aggregatory phenotype in cells capable of accumulating enough cytosolic calcium to trigger specific pathways that mediate the loss of their aggregating properties and the development of new adhesive and procoagulant characteristics. Complex cascades of signaling events are involved and this may explain why an inter-individual variability exists in procoagulant potential. Nowadays, we know the key agonists and mediators underlying the generation of a procoagulant platelet response. However, we still lack insight into the actual mechanisms controlling this dichotomous pattern (i.e., procoagulant versus aggregating phenotype). In this review, we describe the phenotypic characteristics of procoagulant COAT platelets, we detail the current knowledge on the mechanisms of the procoagulant response, and discuss possible drivers of this dichotomous diversification, in particular addressing the impact of the platelet environment during in vivo thrombus formation.
Collapse
Affiliation(s)
| | | | | | | | - Lorenzo Alberio
- Hemostasis and Platelet Research Laboratory, Division of Hematology and Central Hematology Laboratory, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), CH-1010 Lausanne, Switzerland; (L.V.); (A.A.); (D.B.C.); (C.P.P.)
| |
Collapse
|
28
|
Perrella G, Montague SJ, Brown HC, Garcia Quintanilla L, Slater A, Stegner D, Thomas M, Heemskerk JWM, Watson SP. Role of Tyrosine Kinase Syk in Thrombus Stabilisation at High Shear. Int J Mol Sci 2022; 23:ijms23010493. [PMID: 35008919 PMCID: PMC8745592 DOI: 10.3390/ijms23010493] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 12/18/2022] Open
Abstract
Understanding the pathways involved in the formation and stability of the core and shell regions of a platelet-rich arterial thrombus may result in new ways to treat arterial thrombosis. The distinguishing feature between these two regions is the absence of fibrin in the shell which indicates that in vitro flow-based assays over thrombogenic surfaces, in the absence of coagulation, can be used to resemble this region. In this study, we have investigated the contribution of Syk tyrosine kinase in the stability of platelet aggregates (or thrombi) formed on collagen or atherosclerotic plaque homogenate at arterial shear (1000 s-1). We show that post-perfusion of the Syk inhibitor PRT-060318 over preformed thrombi on both surfaces enhances thrombus breakdown and platelet detachment. The resulting loss of thrombus stability led to a reduction in thrombus contractile score which could be detected as early as 3 min after perfusion of the Syk inhibitor. A similar loss of thrombus stability was observed with ticagrelor and indomethacin, inhibitors of platelet adenosine diphosphate (ADP) receptor and thromboxane A2 (TxA2), respectively, and in the presence of the Src inhibitor, dasatinib. In contrast, the Btk inhibitor, ibrutinib, causes only a minor decrease in thrombus contractile score. Weak thrombus breakdown is also seen with the blocking GPVI nanobody, Nb21, which indicates, at best, a minor contribution of collagen to the stability of the platelet aggregate. These results show that Syk regulates thrombus stability in the absence of fibrin in human platelets under flow and provide evidence that this involves pathways additional to activation of GPVI by collagen.
Collapse
Affiliation(s)
- Gina Perrella
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (G.P.); (S.J.M.); (H.C.B.); (L.G.Q.); (A.S.); (M.T.)
- Department of Biochemistry, CARIM, Maastricht University, 6200 AC Maastricht, The Netherlands;
| | - Samantha J. Montague
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (G.P.); (S.J.M.); (H.C.B.); (L.G.Q.); (A.S.); (M.T.)
| | - Helena C. Brown
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (G.P.); (S.J.M.); (H.C.B.); (L.G.Q.); (A.S.); (M.T.)
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, 97080 Würzburg, Germany;
| | - Lourdes Garcia Quintanilla
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (G.P.); (S.J.M.); (H.C.B.); (L.G.Q.); (A.S.); (M.T.)
| | - Alexandre Slater
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (G.P.); (S.J.M.); (H.C.B.); (L.G.Q.); (A.S.); (M.T.)
| | - David Stegner
- Institute of Experimental Biomedicine I, University Hospital, University of Würzburg, 97080 Würzburg, Germany;
| | - Mark Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (G.P.); (S.J.M.); (H.C.B.); (L.G.Q.); (A.S.); (M.T.)
| | - Johan W. M. Heemskerk
- Department of Biochemistry, CARIM, Maastricht University, 6200 AC Maastricht, The Netherlands;
- Department Synapse Research Institute, 6214 AC Maastricht, The Netherlands
| | - Steve P. Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (G.P.); (S.J.M.); (H.C.B.); (L.G.Q.); (A.S.); (M.T.)
- Centre of Membrane Proteins and Receptors (COMPARE), The Universities of Birmingham, Birmingham B15 2TT, UK
- Centre of Membrane Proteins and Receptors (COMPARE), The Universities of Nottingham, Nottingham NG7 2RD, UK
- Correspondence: ; Tel.: +44-0121-4146514
| |
Collapse
|
29
|
Jourdi G, Lordkipanidzé M, Philippe A, Bachelot-Loza C, Gaussem P. Current and Novel Antiplatelet Therapies for the Treatment of Cardiovascular Diseases. Int J Mol Sci 2021; 22:ijms222313079. [PMID: 34884884 PMCID: PMC8658271 DOI: 10.3390/ijms222313079] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/22/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
Over the last decades, antiplatelet agents, mainly aspirin and P2Y12 receptor antagonists, have significantly reduced morbidity and mortality associated with arterial thrombosis. Their pharmacological characteristics, including pharmacokinetic/pharmacodynamics profiles, have been extensively studied, and a significant number of clinical trials assessing their efficacy and safety in various clinical settings have established antithrombotic efficacy. Notwithstanding, antiplatelet agents carry an inherent risk of bleeding. Given that bleeding is associated with adverse cardiovascular outcomes and mortality, there is an unmet clinical need to develop novel antiplatelet therapies that inhibit thrombosis while maintaining hemostasis. In this review, we present the currently available antiplatelet agents, with a particular focus on their targets, pharmacological characteristics, and patterns of use. We will further discuss the novel antiplatelet therapies in the pipeline, with the goal of improved clinical outcomes among patients with atherothrombotic diseases.
Collapse
Affiliation(s)
- Georges Jourdi
- Research Center, Montreal Heart Institute, Montreal, QC H1T 1C8, Canada;
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Correspondence: (G.J.); (P.G.)
| | - Marie Lordkipanidzé
- Research Center, Montreal Heart Institute, Montreal, QC H1T 1C8, Canada;
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Aurélien Philippe
- INSERM, Innovations Thérapeutiques en Hémostase, Université de Paris, F-75006 Paris, France; (A.P.); (C.B.-L.)
- Service d’Hématologie Biologique, AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Christilla Bachelot-Loza
- INSERM, Innovations Thérapeutiques en Hémostase, Université de Paris, F-75006 Paris, France; (A.P.); (C.B.-L.)
| | - Pascale Gaussem
- INSERM, Innovations Thérapeutiques en Hémostase, Université de Paris, F-75006 Paris, France; (A.P.); (C.B.-L.)
- Service d’Hématologie Biologique, AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France
- Correspondence: (G.J.); (P.G.)
| |
Collapse
|
30
|
Ponomarenko EA, Ignatova AA, Polokhov DM, Khismatullina RD, Kurilo DS, Shcherbina A, Zharkov PA, Maschan AA, Novichkova GA, Panteleev MA. Healthy pediatric platelets are moderately hyporeactive in comparison with adults' platelets. Platelets 2021; 33:727-734. [PMID: 34749589 DOI: 10.1080/09537104.2021.1981848] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Studies on platelet function in children older than neonatal period are few and their results are controversial. The pediatric platelets were alternatively reported to be more active or less active than adults' ones. We compared platelet function in the several age groups of children to adults and evaluated the age when platelet function reaches the adults' status. The study included 76 healthy children and 49 healthy adult volunteers. Types of platelet activation used included: collagen-related peptide (CRP) and PAR-1 activating peptide SFLLRN; SFLLRN, PAR-4 activating peptide AYPGKF and adenosine diphosphate (ADP); ADP. The parameters determined included forward (FSC) and side scatter (SSC), CD42b, CD61, CD62P, PAC-1, annexin V binding and mepacrine release levels. Resting pediatric platelets were similar to adults' platelets except for 1.2-fold decreased FSC and dense granules volume in youngest children, and 2.5-fold increased annexin V level in children aged 1-10 years. After CRP+SFLLRN stimulation, pediatric platelets had a 1.2-fold lower alpha- and 1.1-fold lower dense granule release than adults. For SFLLRN+AYPGKF+ADP stimulation, this was observed only for youngest children. The response to ADP stimulation was identical for pediatric platelets and adults. Pediatric platelets have lower granular release than adults' platelets, which persists until the age of 18.
Collapse
Affiliation(s)
- Evgeniya A Ponomarenko
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology Named after Dmitry Rogachev, Russian Ministry of Healthcare, Cellular Hemostasis and Thrombosis Lab, Moscow, Russian Federation.,M.V. Lomonosov Moscow State University, Faculty of Biology, Moscow, Russian Federation
| | - Anastasia A Ignatova
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology Named after Dmitry Rogachev, Russian Ministry of Healthcare, Cellular Hemostasis and Thrombosis Lab, Moscow, Russian Federation.,Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Laboratory of Molecular Mechanisms of Hemostasis, Moscow, Russian Federation
| | - Dmitrii M Polokhov
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology Named after Dmitry Rogachev, Russian Ministry of Healthcare, Cellular Hemostasis and Thrombosis Lab, Moscow, Russian Federation
| | - Rimma D Khismatullina
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology Named after Dmitry Rogachev, Russian Ministry of Healthcare, Cellular Hemostasis and Thrombosis Lab, Moscow, Russian Federation
| | - Darja S Kurilo
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology Named after Dmitry Rogachev, Russian Ministry of Healthcare, Cellular Hemostasis and Thrombosis Lab, Moscow, Russian Federation
| | - Anna Shcherbina
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology Named after Dmitry Rogachev, Russian Ministry of Healthcare, Cellular Hemostasis and Thrombosis Lab, Moscow, Russian Federation
| | - Pavel A Zharkov
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology Named after Dmitry Rogachev, Russian Ministry of Healthcare, Cellular Hemostasis and Thrombosis Lab, Moscow, Russian Federation
| | - Alexey A Maschan
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology Named after Dmitry Rogachev, Russian Ministry of Healthcare, Cellular Hemostasis and Thrombosis Lab, Moscow, Russian Federation
| | - Galina A Novichkova
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology Named after Dmitry Rogachev, Russian Ministry of Healthcare, Cellular Hemostasis and Thrombosis Lab, Moscow, Russian Federation
| | - Mikhail A Panteleev
- National Medical Research Center of Pediatric Hematology, Oncology and Immunology Named after Dmitry Rogachev, Russian Ministry of Healthcare, Cellular Hemostasis and Thrombosis Lab, Moscow, Russian Federation.,Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Laboratory of Molecular Mechanisms of Hemostasis, Moscow, Russian Federation.,M.V. Lomonosov Moscow State University, Faculty of Physics, Moscow, Russian Federation
| |
Collapse
|
31
|
Chaudhary PK, Kim S, Kim S. The Predominant Role of Arrestin3 in General GPCR Desensitization in Platelets. J Clin Med 2021; 10:jcm10204743. [PMID: 34682866 PMCID: PMC8539091 DOI: 10.3390/jcm10204743] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 01/14/2023] Open
Abstract
Arrestins in concert with GPCR kinases (GRKs) function in G protein-coupled receptor (GPCR) desensitization in various cells. Therefore, we characterized the functional differences of arrestin3 versus arrestin2 in the regulation of GPCR signaling and its desensitization in platelets using mice lacking arrestin3 and arrestin2. In contrast to arrestin2, platelet aggregation and dense granule secretion induced by 2-MeSADP, U46619, thrombin, and AYPGKF were significantly potentiated in arrestin3-deficient platelets compared to wild-type (WT) platelets, while non-GPCR agonist CRP-induced platelet aggregation and secretion were not affected. Surprisingly, in contrast to GRK6, platelet aggregation induced by the co-stimulation of serotonin and epinephrine was significantly potentiated in arrestin3-deficient platelets, suggesting the central role of arrestin3 in general GPCR desensitization in platelets. In addition, the second challenge of ADP and AYPGKF restored platelet aggregation in arrestin3-deficient platelets but failed to do so in WT and arrestin2-deficient platelets, confirming that arrestin3 contributes to GPCR desensitization. Furthermore, ADP- and AYPGKF-induced Akt and ERK phosphorylation were significantly increased in arrestin3-deficient platelets. Finally, we found that arrestin3 is critical for thrombus formation in vivo. In conclusion, arrestin3, not arrestin2, plays a central role in the regulation of platelet functional responses and thrombus formation through general GPCR desensitization in platelets.
Collapse
|
32
|
The brain-derived neurotrophic factor prompts platelet aggregation and secretion. Blood Adv 2021; 5:3568-3580. [PMID: 34546355 DOI: 10.1182/bloodadvances.2020004098] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 06/08/2021] [Indexed: 12/19/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) has both autocrine and paracrine roles in neurons, and its release and signaling mechanisms have been extensively studied in the central nervous system. Large quantities of BDNF have been reported in circulation, essentially stored in platelets with concentrations reaching 100- to 1000-fold those of neurons. Despite this abundance, the function of BDNF in platelet biology has not been explored. At low concentrations, BDNF primed platelets, acting synergistically with classical agonists. At high concentrations, BDNF induced complete biphasic platelet aggregation that in part relied on amplification from secondary mediators. Neurotrophin-4, but not nerve growth factor, and an activating antibody against the canonical BDNF receptor tropomyosin-related kinase B (TrkB) induced similar platelet responses to BDNF, suggesting TrkB could be the mediator. Platelets expressed, both at their surface and in their intracellular compartment, a truncated form of TrkB lacking its tyrosine kinase domain. BDNF-induced platelet aggregation was prevented by inhibitors of Ras-related C3 botulinum toxin substrate 1 (Rac1), protein kinase C, and phosphoinositide 3-kinase. BDNF-stimulated platelets secreted a panel of angiogenic and inflammatory cytokines, which may play a role in maintaining vascular homeostasis. Two families with autism spectrum disorder were found to carry rare missense variants in the BDNF gene. Platelet studies revealed defects in platelet aggregation to low concentrations of collagen, as well as reduced adenosine triphosphate secretion in response to adenosine diphosphate. In summary, circulating BDNF levels appear to regulate platelet activation, aggregation, and secretion through activation of a truncated TrkB receptor and downstream kinase-dependent signaling.
Collapse
|
33
|
Pallini C, Pike JA, O'Shea C, Andrews RK, Gardiner EE, Watson SP, Poulter NS. Immobilized collagen prevents shedding and induces sustained GPVI clustering and signaling in platelets. Platelets 2021; 32:59-73. [PMID: 33455536 DOI: 10.1080/09537104.2020.1849607] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Collagen, the most thrombogenic constituent of blood vessel walls, activates platelets through glycoprotein VI (GPVI). In suspension, following platelet activation by collagen, GPVI is cleaved by A Disintegrin And Metalloproteinase (ADAM)10 and ADAM17. In this study, we use single-molecule localization microscopy and a 2-level DBSCAN-based clustering tool to show that GPVI remains clustered along immobilized collagen fibers for at least 3 hours in the absence of significant shedding. Tyrosine phosphorylation of spleen tyrosine kinase (Syk) and Linker of Activated T cells (LAT), and elevation of intracellular Ca2+, are sustained over this period. Syk, but not Src kinase-dependent signaling is required to maintain clustering of the collagen integrin α2β1, whilst neither is required for GPVI. We propose that clustering of GPVI on immobilized collagen protects GPVI from shedding in order to maintain sustained Src and Syk-kinases dependent signaling, activation of integrin α2β1, and continued adhesion.
Collapse
Affiliation(s)
- Chiara Pallini
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham , Midlands, UK
| | - Jeremy A Pike
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - Christopher O'Shea
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - Robert K Andrews
- Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, Australian National University , Canberra, Australia
| | - Elizabeth E Gardiner
- Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, Australian National University , Canberra, Australia
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham , Midlands, UK
| | - Natalie S Poulter
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham , Midlands, UK
| |
Collapse
|
34
|
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
|
35
|
Bleeding diathesis in mice lacking JAK2 in platelets. Blood Adv 2021; 5:2969-2981. [PMID: 34342643 DOI: 10.1182/bloodadvances.2020003032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 04/14/2021] [Indexed: 12/18/2022] Open
Abstract
The tyrosine kinase JAK2 is a critical component of intracellular JAK/STAT cytokine signaling cascades that is prevalent in hematopoietic cells, such as hematopoietic stem cells and megakaryocytes (MKs). Individuals expressing the somatic JAK2 V617F mutation commonly develop myeloproliferative neoplasms (MPNs) associated with venous and arterial thrombosis, a leading cause of mortality. The role of JAK2 in hemostasis remains unclear. We investigated the role of JAK2 in platelet hemostatic function using Jak2fl/fl Pf4-Cre (Jak2Plt-/-) mice lacking JAK2 in platelets and MKs. Jak2Plt-/- mice developed MK hyperplasia and splenomegaly associated with severe thrombocytosis and bleeding. This notion was supported by failure to occlude in a ferric chloride carotid artery injury model and by a cremaster muscle laser-induced injury assay, in which Jak2Plt-/- platelets failed to form stable thrombi. Jak2Plt-/- platelets formed thrombi poorly after adhesion to type 1 collagen under arterial shear rates. Jak2Plt-/- platelets spread poorly on collagen under static conditions or on fibrinogen in response to the collagen receptor GPVI-specific agonist, collagen-related peptide (CRP). After activation with collagen, CRP, or the CLEC-2 agonist rhodocytin, Jak2Plt-/- platelets displayed decreased α-granule secretion and integrin αIIbβ3 activation or aggregation, but showed normal responses to thrombin. Jak2Plt-/- platelets had impaired intracellular signaling when activated via GPVI, as assessed by tyrosine phosphorylation. Together, the results show that JAK2 deletion impairs platelet immunoreceptor tyrosine-based activation motif signaling and hemostatic function in mice and suggest that aberrant JAK2 signaling in patients with MPNs affects GPVI signaling, leading to hemostatic platelet function.
Collapse
|
36
|
Liu G, Yuan Z, Tian X, Xiong X, Guo F, Lin Z, Qin Z. Pimpinellin Inhibits Collagen-induced Platelet Aggregation and Activation Through Inhibiting Granule Secretion and PI3K/Akt Pathway. Front Pharmacol 2021; 12:706363. [PMID: 34366861 PMCID: PMC8339208 DOI: 10.3389/fphar.2021.706363] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/07/2021] [Indexed: 01/21/2023] Open
Abstract
Pimpinellin is a coumarin-like compound extracted from the root of Toddalia asiatica. Its effects on platelet function has not been investigated. This study found that pimpinellin pretreatment effectively inhibited collagen-induced platelet aggregation, but did not alter ADP- and thrombin-induced aggregation. Platelets pretreated with pimpinellin showed reduced α granule (CD62) level and secretion of dense granule (ATP release). Pimpinellin-treated platelets also exhibited decreased clot reaction and TxB2 production. Pimpinellin pretreatment suppressed adhesion and spreading of human platelets on the fibrinogen coated surface. Analysis of tail bleeding time of mice administered with pimpinellin (40 mg/kg) revealed that pimpinellin did not change tail bleeding time significantly, number of blood cells, and APTT and PT levels. Pimpinellin inhibited collagen-induced ex vivo aggregation of mice platelets. Immunoblotting results showed that pimpinellin suppressed collagen-induced phosphorylation of PI3K-Akt-Gsk3β and PKC/MAPK in platelets.
Collapse
Affiliation(s)
- Gang Liu
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
| | - Zhaowei Yuan
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Xiaoyun Tian
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Xiuqin Xiong
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Fang Guo
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Zihan Lin
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, China
| | - Zhen Qin
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| |
Collapse
|
37
|
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
|
38
|
Tao DL, Tassi Yunga S, Williams CD, McCarty OJT. Aspirin and antiplatelet treatments in cancer. Blood 2021; 137:3201-3211. [PMID: 33940597 PMCID: PMC8351882 DOI: 10.1182/blood.2019003977] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
Platelets have been hypothesized to promote certain neoplastic malignancies; however, antiplatelet drugs are still not part of routine pharmacological cancer prevention and treatment protocols. Paracrine interactions between platelets and cancer cells have been implicated in potentiating the dissemination, survival within the circulation, and extravasation of cancer cells at distant sites of metastasis. Signals from platelets have also been suggested to confer epigenetic alterations, including upregulating oncoproteins in circulating tumor cells, and secretion of potent growth factors may play roles in promoting mitogenesis, angiogenesis, and metastatic outgrowth. Thrombocytosis remains a marker of poor prognosis in patients with solid tumors. Experimental data suggest that lowering of platelet count may reduce tumor growth and metastasis. On the basis of the mechanisms by which platelets could contribute to cancer growth and metastasis, it is conceivable that drugs reducing platelet count or platelet activation might attenuate cancer progression and improve outcomes. We will review select pharmacological approaches that inhibit platelets and may affect cancer development and propagation. We begin by presenting an overview of clinical cancer prevention and outcome studies with low-dose aspirin. We then review current nonclinical development of drugs targeted to platelet binding, activation, and count as potential mitigating agents in cancer.
Collapse
Affiliation(s)
- Derrick L Tao
- Division of Hematology & Medical Oncology
- Department of Biomedical Engineering, and
| | - Samuel Tassi Yunga
- Department of Biomedical Engineering, and
- Cancer Early Detection & Advanced Research Center, Oregon Health & Science University, Portland, OR; and
| | - Craig D Williams
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, OR
| | - Owen J T McCarty
- Division of Hematology & Medical Oncology
- Department of Biomedical Engineering, and
| |
Collapse
|
39
|
Zheng TJ, Lofurno ER, Melrose AR, Lakshmanan HHS, Pang J, Phillips KG, Fallon ME, Kohs TCL, Ngo ATP, Shatzel JJ, Hinds MT, McCarty OJT, Aslan JE. Assessment of the effects of Syk and BTK inhibitors on GPVI-mediated platelet signaling and function. Am J Physiol Cell Physiol 2021; 320:C902-C915. [PMID: 33689480 PMCID: PMC8163578 DOI: 10.1152/ajpcell.00296.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/25/2022]
Abstract
Spleen tyrosine kinase (Syk) and Bruton's tyrosine kinase (BTK) play critical roles in platelet physiology, facilitating intracellular immunoreceptor tyrosine-based activation motif (ITAM)-mediated signaling downstream of platelet glycoprotein VI (GPVI) and GPIIb/IIIa receptors. Small molecule tyrosine kinase inhibitors (TKIs) targeting Syk and BTK have been developed as antineoplastic and anti-inflammatory therapeutics and have also gained interest as antiplatelet agents. Here, we investigate the effects of 12 different Syk and BTK inhibitors on GPVI-mediated platelet signaling and function. These inhibitors include four Syk inhibitors, Bay 61-3606, R406 (fostamatinib), entospletinib, TAK-659; four irreversible BTK inhibitors, ibrutinib, acalabrutinib, ONO-4059 (tirabrutinib), AVL-292 (spebrutinib); and four reversible BTK inhibitors, CG-806, BMS-935177, BMS-986195, and fenebrutinib. In vitro, TKIs targeting Syk or BTK reduced platelet adhesion to collagen, dense granule secretion, and alpha granule secretion in response to the GPVI agonist cross-linked collagen-related peptide (CRP-XL). Similarly, these TKIs reduced the percentage of activated integrin αIIbβ3 on the platelet surface in response to CRP-XL, as determined by PAC-1 binding. Although all TKIs tested inhibited phospholipase C γ2 (PLCγ2) phosphorylation following GPVI-mediated activation, other downstream signaling events proximal to phosphoinositide 3-kinase (PI3K) and PKC were differentially affected. In addition, reversible BTK inhibitors had less pronounced effects on GPIIb/IIIa-mediated platelet spreading on fibrinogen and differentially altered the organization of PI3K around microtubules during platelets spreading on fibrinogen. Select TKIs also inhibited platelet aggregate formation on collagen under physiological flow conditions. Together, our results suggest that TKIs targeting Syk or BTK inhibit central platelet functional responses but may differentially affect protein activities and organization in critical systems downstream of Syk and BTK in platelets.
Collapse
Affiliation(s)
- Tony J Zheng
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Elizabeth R Lofurno
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Alexander R Melrose
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | | | - Jiaqing Pang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | | | - Meghan E Fallon
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Tia C L Kohs
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Anh T P Ngo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Joseph J Shatzel
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Monica T Hinds
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Joseph E Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
- Department of Chemical Physiology and Biochemistry, School of Medicine, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
40
|
Shu D, Zhu Y, Lu M, He AD, Chen JB, Ye DS, Liu Y, Zeng XB, Ma R, Ming ZY. Sanguinarine Attenuates Collagen-Induced Platelet Activation and Thrombus Formation. Biomedicines 2021; 9:biomedicines9050444. [PMID: 33919019 PMCID: PMC8142988 DOI: 10.3390/biomedicines9050444] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 11/16/2022] Open
Abstract
Sanguinarine, a benzophenanthridine alkaloid, has been described to have an antiplatelet activity. However, its antithrombotic effect and the mechanism of platelet inhibition have not thoroughly been explored. The current study found that sanguinarine had an inhibitory effect on thrombus formation. This inhibitory effect was quite evident both in the flow-chamber assays as well as in a murine model of FeCl3-induced carotid artery thrombosis. Further investigations also revealed that sanguinarine inhibited the collagen-induced human platelet aggregation and granule release. At the same time, it also prevented platelet spreading and adhesion to immobilized fibrinogen. The molecular mechanisms of its antiplatelet activity were found to be as follows: 1. Reduced phosphorylation of the downstream signaling pathways in collagen specific receptor GPVI (Syk-PLCγ2 and PI3K-Akt-GSK3β); 2. Inhibition of collagen-induced increase in the intracellular Ca2+ concentration ([Ca2+]i); 3. Inhibition of integrin αIIbβ3 outside-in signaling via reducing β3 and Src (Tyr-416) phosphorylation. It can be concluded that sanguinarine inhibits collagen-induced platelet activation and reduces thrombus formation. This effect is mediated via inhibiting the phosphorylation of multiple components in the GPVI signaling pathway. Current data also indicate that sanguinarine can be of some clinical value to treat cardiovascular diseases involving an excess of platelet activation.
Collapse
Affiliation(s)
- Dan Shu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong, University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China; (D.S.); (Y.Z.); (M.L.); (J.-B.C.); (D.-S.Y.); (Y.L.); (X.-B.Z.); (R.M.)
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, 13 Hangkong Road, Wuhan 430030, China
- College of Pharmacy, Xiangnan University, 889 Chenzhou Avenue, Chenzhou 423000, China
| | - Ying Zhu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong, University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China; (D.S.); (Y.Z.); (M.L.); (J.-B.C.); (D.-S.Y.); (Y.L.); (X.-B.Z.); (R.M.)
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, 13 Hangkong Road, Wuhan 430030, China
| | - Meng Lu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong, University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China; (D.S.); (Y.Z.); (M.L.); (J.-B.C.); (D.-S.Y.); (Y.L.); (X.-B.Z.); (R.M.)
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, 13 Hangkong Road, Wuhan 430030, China
| | - Ao-Di He
- Department of Physiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan 430030, China;
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiang-Bin Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong, University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China; (D.S.); (Y.Z.); (M.L.); (J.-B.C.); (D.-S.Y.); (Y.L.); (X.-B.Z.); (R.M.)
| | - Ding-Song Ye
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong, University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China; (D.S.); (Y.Z.); (M.L.); (J.-B.C.); (D.-S.Y.); (Y.L.); (X.-B.Z.); (R.M.)
| | - Yue Liu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong, University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China; (D.S.); (Y.Z.); (M.L.); (J.-B.C.); (D.-S.Y.); (Y.L.); (X.-B.Z.); (R.M.)
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, 13 Hangkong Road, Wuhan 430030, China
| | - Xiang-Bin Zeng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong, University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China; (D.S.); (Y.Z.); (M.L.); (J.-B.C.); (D.-S.Y.); (Y.L.); (X.-B.Z.); (R.M.)
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, 13 Hangkong Road, Wuhan 430030, China
| | - Rong Ma
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong, University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China; (D.S.); (Y.Z.); (M.L.); (J.-B.C.); (D.-S.Y.); (Y.L.); (X.-B.Z.); (R.M.)
| | - Zhang-Yin Ming
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong, University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China; (D.S.); (Y.Z.); (M.L.); (J.-B.C.); (D.-S.Y.); (Y.L.); (X.-B.Z.); (R.M.)
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, 13 Hangkong Road, Wuhan 430030, China
- Tongji-Rongcheng Center for Biomedicine, Huazhong University of Science and Technology, Wuhan 430030, China
- Correspondence: ; Tel.: +86-27-83650710
| |
Collapse
|
41
|
von Hundelshausen P, Siess W. Bleeding by Bruton Tyrosine Kinase-Inhibitors: Dependency on Drug Type and Disease. Cancers (Basel) 2021; 13:1103. [PMID: 33806595 PMCID: PMC7961939 DOI: 10.3390/cancers13051103] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
Bruton tyrosine kinase (Btk) is expressed in B-lymphocytes, myeloid cells and platelets, and Btk-inhibitors (BTKi) are used to treat patients with B-cell malignancies, developed against autoimmune diseases, have been proposed as novel antithrombotic drugs, and been tested in patients with severe COVID-19. However, mild bleeding is frequent in patients with B-cell malignancies treated with the irreversible BTKi ibrutinib and the recently approved 2nd generation BTKi acalabrutinib, zanubrutinib and tirabrutinib, and also in volunteers receiving in a phase-1 study the novel irreversible BTKi BI-705564. In contrast, no bleeding has been reported in clinical trials of other BTKi. These include the brain-penetrant irreversible tolebrutinib and evobrutinib (against multiple sclerosis), the irreversible branebrutinib, the reversible BMS-986142 and fenebrutinib (targeting rheumatoid arthritis and lupus erythematodes), and the reversible covalent rilzabrutinib (against pemphigus and immune thrombocytopenia). Remibrutinib, a novel highly selective covalent BTKi, is currently in clinical studies of autoimmune dermatological disorders. This review describes twelve BTKi approved or in clinical trials. By focusing on their pharmacological properties, targeted disease, bleeding side effects and actions on platelets it attempts to clarify the mechanisms underlying bleeding. Specific platelet function tests in blood might help to estimate the probability of bleeding of newly developed BTKi.
Collapse
Affiliation(s)
- Philipp von Hundelshausen
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University (LMU), 80336 Munich, Germany;
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Wolfgang Siess
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University (LMU), 80336 Munich, Germany;
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| |
Collapse
|
42
|
Abstract
Platelets are the major cellular contributor to arterial thrombosis. However, activated platelets form two distinct subpopulations during thrombosis. Pro-aggregatory platelets aggregate to form the main body of the thrombus. In contrast, procoagulant platelets expose phosphatidylserine on their outer surface and promote thrombin generation. This apparently all-or-nothing segregation into subpopulations indicates that, during activation, platelets commit to becoming procoagulant or pro-aggregatory. Although the signaling pathways that control this commitment are not understood, distinct cytosolic and mitochondrial Ca2+ signals in different subpopulations are likely to be central. In this review, we discuss how these Ca2+ signals control procoagulant platelet formation and whether this process can be targeted pharmacologically to prevent arterial thrombosis.
Collapse
Affiliation(s)
| | - Matthew T Harper
- Department of Pharmacology, University of Cambridge Cambridge, UK
| |
Collapse
|
43
|
Droplet Microfluidics with Reagent Micromixing for Investigating Intrinsic Platelet Functionality. Cell Mol Bioeng 2021; 14:223-230. [PMID: 34109001 DOI: 10.1007/s12195-020-00665-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/23/2020] [Indexed: 10/22/2022] Open
Abstract
Introduction Precision mapping of the functional structure of platelet populations holds great promise for the identification of hyper-reactive subtypes that are likely to be disease drivers, having value in prognostics and as therapeutic targets. However, the ability to measure the intrinsic functional capacity of individual platelets is confounded by potent paracrine cross-talk, resulting in phenotypic remodeling of the entire platelet population, and in doing so obscuring the identity of hyper-reactive platelets. Methods To address this we have developed a droplet microfluidics strategy for single platelet confinement to exclude paracrine signaling. Consideration of the Poisson distribution was used for high throughput single platelet encapsulation and the preparation of minimal platelet collectives serving as digital models for understanding the role of hyper-reactive platelets coordinating system-level behavior by paracrine signaling. Platelets are retrieved from the droplets for phenotyping using standard flow cytometry. In addition, we have incorporated a staggered herringbone micromixing element for accurate agonist and antibody dispensing in droplets. Results The methodology was used for characterizing sensitivity distributions from healthy blood donors in response to convulxin (agonist of the GPVI receptor, the major platelet receptor for collagen). P-selectin exposure and α IIb β 3 integrin activation were used as analytical end-points to demonstrate the existence of hyper-reactive platelets that direct 20-fold gains in system level sensitivity. Conclusions The analytical workflow represents an enabling tool for the accurate classification of platelet subtypes and description of their underlying biology. Supplementary information The online version of this article (10.1007/s12195-020-00665-6) contains supplementary material, which is available to authorized users.
Collapse
|
44
|
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
|
45
|
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]
|
46
|
Harbi MH, Smith CW, Nicolson PLR, Watson SP, Thomas MR. Novel antiplatelet strategies targeting GPVI, CLEC-2 and tyrosine kinases. Platelets 2020; 32:29-41. [PMID: 33307909 DOI: 10.1080/09537104.2020.1849600] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Antiplatelet medications comprise the cornerstone of treatment for diseases that involve arterial thrombosis, including acute coronary syndromes (ACS), stroke and peripheral arterial disease. However, antiplatelet medications may cause bleeding and, furthermore, thrombotic events may still recur despite treatment. The interaction of collagen with GPVI receptors on the surface of platelets has been identified as one of the major players in the pathophysiology of arterial thrombosis that occurs following atherosclerotic plaque rupture. Promisingly, GPVI deficiency in humans appears to have a minimal impact on bleeding. These findings together suggest that targeting platelet GPVI may provide a novel treatment strategy that provides additional antithrombotic efficacy with minimal disruption of normal hemostasis compared to conventional antiplatelet medications. CLEC-2 is gaining interest as a therapeutic target for a variety of thrombo-inflammatory disorders including deep vein thrombosis (DVT) with treatment also predicted to cause minimal disruption to hemostasis. GPVI and CLEC-2 signal through Src, Syk and Tec family tyrosine kinases, providing additional strategies for inhibiting both receptors. In this review, we summarize the evidence regarding GPVI and CLEC-2 and strategies for inhibiting these receptors to inhibit platelet recruitment and activation in thrombotic diseases.
Collapse
Affiliation(s)
- Maan H Harbi
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - Christopher W Smith
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - Phillip L R Nicolson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK.,University Hospitals Birmingham NHS Foundation Trust , Birmingham, UK
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK
| | - Mark R Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham , Birmingham, UK.,University Hospitals Birmingham NHS Foundation Trust , Birmingham, UK.,Sandwell and West Birmingham NHS Trust , Birmingham, UK
| |
Collapse
|
47
|
Perrella G, Huang J, Provenzale I, Swieringa F, Heubel-Moenen FCJI, Farndale RW, Roest M, van der Meijden PEJ, Thomas M, Ariëns RAS, Jandrot-Perrus M, Watson SP, Heemskerk JWM. Nonredundant Roles of Platelet Glycoprotein VI and Integrin αIIbβ3 in Fibrin-Mediated Microthrombus Formation. Arterioscler Thromb Vasc Biol 2020; 41:e97-e111. [PMID: 33267658 DOI: 10.1161/atvbaha.120.314641] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Fibrin is considered to strengthen thrombus formation via integrin αIIbβ3, but recent findings indicate that fibrin can also act as ligand for platelet glycoprotein VI. Approach and Results: To investigate the thrombus-forming potential of fibrin and the roles of platelet receptors herein, we generated a range of immobilized fibrin surfaces, some of which were cross-linked with factor XIIIa and contained VWF-BP (von Willebrand factor-binding peptide). Multicolor microfluidics assays with whole-blood flowed at high shear rate (1000 s-1) indicated that the fibrin surfaces, regardless of the presence of factor XIIIa or VWF-BP, supported platelet adhesion and activation (P-selectin expression), but only microthrombi were formed consisting of bilayers of platelets. Fibrinogen surfaces produced similar microthrombi. Markedly, tiggering of coagulation with tissue factor or blocking of thrombin no more than moderately affected the fibrin-induced microthrombus formation. Absence of αIIbβ3 in Glanzmann thrombasthenia annulled platelet adhesion. Blocking of glycoprotein VI with Fab 9O12 substantially, but incompletely reduced platelet secretion, Ca2+ signaling and aggregation, while inhibition of Syk further reduced these responses. In platelet suspension, glycoprotein VI blockage or Syk inhibition prevented fibrin-induced platelet aggregation. Microthrombi on fibrin surfaces triggered only minimal thrombin generation, in spite of thrombin binding to the fibrin fibers. CONCLUSIONS Together, these results indicate that fibrin fibers, regardless of their way of formation, act as a consolidating surface in microthrombus formation via nonredundant roles of platelet glycoprotein VI and integrin αIIbβ3 through signaling via Syk and low-level Ca2+ rises.
Collapse
Affiliation(s)
- Gina Perrella
- Department of Biochemistry, CARIM, Maastricht University, the Netherlands (G.P., J.H., I.P., F.S., P.E.J.v.d.M., R.A.S.A., S.P.W., J.W.M.H.)
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (G.P., M.T., S.P.W.)
| | - Jingnan Huang
- Department of Biochemistry, CARIM, Maastricht University, the Netherlands (G.P., J.H., I.P., F.S., P.E.J.v.d.M., R.A.S.A., S.P.W., J.W.M.H.)
- ISAS Institute, Dortmund, DE (J.H.)
| | - Isabella Provenzale
- Department of Biochemistry, CARIM, Maastricht University, the Netherlands (G.P., J.H., I.P., F.S., P.E.J.v.d.M., R.A.S.A., S.P.W., J.W.M.H.)
| | - Frauke Swieringa
- Department of Biochemistry, CARIM, Maastricht University, the Netherlands (G.P., J.H., I.P., F.S., P.E.J.v.d.M., R.A.S.A., S.P.W., J.W.M.H.)
| | | | - Richard W Farndale
- Department of Biochemistry, University of Cambridge, United Kingdom (R.W.F.)
| | - Mark Roest
- Synapse Research Institute, Maastricht, the Netherlands (M.R.)
| | - Paola E J van der Meijden
- Department of Biochemistry, CARIM, Maastricht University, the Netherlands (G.P., J.H., I.P., F.S., P.E.J.v.d.M., R.A.S.A., S.P.W., J.W.M.H.)
| | - Mark Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (G.P., M.T., S.P.W.)
| | - Robert A S Ariëns
- Department of Biochemistry, CARIM, Maastricht University, the Netherlands (G.P., J.H., I.P., F.S., P.E.J.v.d.M., R.A.S.A., S.P.W., J.W.M.H.)
- Department of Discovery and Translational Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (R.A.S.A.)
| | - Martine Jandrot-Perrus
- UMR S1148, Laboratory for Vascular Translational Science, INSERM, University Paris Diderot, France (M.J.-P.)
| | - Steve P Watson
- Department of Biochemistry, CARIM, Maastricht University, the Netherlands (G.P., J.H., I.P., F.S., P.E.J.v.d.M., R.A.S.A., S.P.W., J.W.M.H.)
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (G.P., M.T., S.P.W.)
- COMPARE, The Universities of Birmingham and Nottingham, the Midlands, United Kingdom (S.P.W.)
| | - Johan W M Heemskerk
- Department of Biochemistry, CARIM, Maastricht University, the Netherlands (G.P., J.H., I.P., F.S., P.E.J.v.d.M., R.A.S.A., S.P.W., J.W.M.H.)
| |
Collapse
|
48
|
Series J, Ribes A, Garcia C, Souleyreau P, Bauters A, Morschhauser F, Jürgensmeier JM, Sié P, Ysebaert L, Payrastre B. Effects of novel Btk and Syk inhibitors on platelet functions alone and in combination in vitro and in vivo. J Thromb Haemost 2020; 18:3336-3351. [PMID: 32926549 DOI: 10.1111/jth.15098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 07/25/2020] [Accepted: 08/31/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Inhibitors of tyrosine kinases downstream of the B-cell receptor, such as Bruton's tyrosine kinase (Btk) or Spleen tyrosine kinase (Syk), used alone or in combination are new therapeutic options in the treatment of B-cell malignancies. A challenge in the development of second-generation Btk inhibitors is to limit their side effects such as the increased bleeding risk. Considering the pivotal role of Syk in immunoreceptor tyrosine-based activation motif mediated platelet signaling, the impact of inhibiting this kinase on platelet functions is also worth analyzing. OBJECTIVES We investigated the effect of a novel Btk inhibitor, tirabrutinib, and a Syk inhibitor, entospletinib, alone and in combination on platelet signaling and functions in vitro and ex vivo. METHODS Platelet aggregation, secretion, and signaling responses as well as thrombus growth under flow were analyzed in the presence of the inhibitors alone or in combination in vitro, at clinically relevant doses, and ex vivo in patients treated with these inhibitors in the context of a phase I trial. RESULTS Although tirabrutinib alone had modest effects on platelet activation in vitro and ex vivo, entospletinib alone efficiently inhibited washed platelet aggregation in response to collagen. However, entospletinib weakly affected platelet activation in platelet-rich plasma, in whole blood and ex vivo. Importantly, the combination of tirabrutinib and entospletinib induced a significant decrease in platelet response to collagen in vitro and ex vivo correlating with mild bleedings reported in some of the treated patients. CONCLUSION These new results should contribute to improve the safety of these targeted therapies.
Collapse
Affiliation(s)
- Jennifer Series
- Inserm, U1048, Université Toulouse 3, I2MC, Toulouse Cedex 04, France
- Laboratoire d'Hématologie CHU de Toulouse, Toulouse Cedex 04, France
| | - Agnès Ribes
- Inserm, U1048, Université Toulouse 3, I2MC, Toulouse Cedex 04, France
- Laboratoire d'Hématologie CHU de Toulouse, Toulouse Cedex 04, France
| | - Cédric Garcia
- Inserm, U1048, Université Toulouse 3, I2MC, Toulouse Cedex 04, France
- Laboratoire d'Hématologie CHU de Toulouse, Toulouse Cedex 04, France
| | - Pierre Souleyreau
- Laboratoire d'Hématologie CHU de Toulouse, Toulouse Cedex 04, France
| | - Anne Bauters
- Institut d'hématologie-transfusion, Laboratoire d'hémostase, CHU Lille, Lille, France
| | | | | | - Pierre Sié
- Inserm, U1048, Université Toulouse 3, I2MC, Toulouse Cedex 04, France
- Laboratoire d'Hématologie CHU de Toulouse, Toulouse Cedex 04, France
| | - Loïc Ysebaert
- Service d'Hématologie IUCT-oncopôle, Toulouse Cedex 09, France
| | - Bernard Payrastre
- Inserm, U1048, Université Toulouse 3, I2MC, Toulouse Cedex 04, France
- Laboratoire d'Hématologie CHU de Toulouse, Toulouse Cedex 04, France
| |
Collapse
|
49
|
Pan G, Chang L, Zhang J, Liu Y, Hu L, Zhang S, Zhang J, Qiao J, Jakopin Ž, Hu H, Dong J, Ding Z. GSK669, a NOD2 receptor antagonist, inhibits thrombosis and oxidative stress via targeting platelet GPVI. Biochem Pharmacol 2020; 183:114315. [PMID: 33152345 DOI: 10.1016/j.bcp.2020.114315] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/21/2020] [Accepted: 10/29/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE Previously, we discovered that the activation of nucleotide-binding oligomerization domain 2 (NOD2) enhances platelet activation. We here investigated the antiplatelet and antithrombotic potential of GSK669, a NOD2 antagonist. EXPERIMENTAL APPROACH Effects of GSK669 on platelet functions, reactive oxygen species (ROS) and proinflammatory cytokine generation were detected. NOD2-/- platelets were used to confirm GSK669 target. The interaction between GSK669 and glycoprotein VI (GPVI) was detected using surface plasmon resonance (SPR) spectroscopy. GPVI downstream signaling was examined by Western blot. The antithrombotic and antioxidative effects were investigated using mouse mesenteric arteriole thrombosis model and pulmonary embolism model. KEY RESULTS GSK669 significantly inhibits platelet proinflammatory cytokine release induced by muramyl dipeptide, platelet aggregation, ATP release, and ROS generation induced by collagen and collagen related peptide (CRP). Platelet spreading and clot retraction are also inhibited. GSK669 also decreases collagen-induced phosphorylation of Src, Syk, PLCγ2, and Akt. The antiplatelet effect of GSK669 is NOD2-independent and mediated by GPVI antagonism. Consistent with its antiplatelet activity as a GPVI antagonist, GSK669 inhibits platelet adhesion on collagen in flow condition. Notably, GSK669 inhibits mouse mesenteric arteriole thrombosis similarly to aspirin without bleeding. The antithrombotic effect of GSK669 is further confirmed in the pulmonary embolism model; decreased malonaldehyde (MDA) and increased superoxide dismutase (SOD) levels in mouse plasma reveal a significant antioxidant effect of GSK669. CONCLUSION AND IMPLICATIONS Beyond its anti-inflammatory effect as a NOD2 antagonist, GSK669 is also an efficient and safe antiplatelet agent combined with antioxidant effect by targeting GPVI. An antiplatelet agent bearing antioxidative and anti-inflammatory activities without bleeding risk may have therapeutic advantage over current antiplatelet drugs for atherothrombosis.
Collapse
Affiliation(s)
- Guanxing Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Lin Chang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jianjun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yangyang Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450018, China
| | - Liang Hu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450018, China
| | - Si Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jian Zhang
- Department of Pathophysiology, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, China
| | - Jianlin Qiao
- Department of Hematology, Blood Disease Institute, Xuzhou Medical University, Xuzhou 221004, China
| | - Žiga Jakopin
- Faculty of Pharmacy, University of Ljubljana, Askerceva 7, SI-1000 Ljubljana, Slovenia
| | - Hu Hu
- Department of Pathophysiology, Zhejiang University School of Medicine, Hangzhou 310012, China
| | - Jianzeng Dong
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450018, China
| | - Zhongren Ding
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450018, China
| |
Collapse
|
50
|
GPR56/ADGRG1 is a platelet collagen-responsive GPCR and hemostatic sensor of shear force. Proc Natl Acad Sci U S A 2020; 117:28275-28286. [PMID: 33097663 PMCID: PMC7668045 DOI: 10.1073/pnas.2008921117] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We identified the known collagen receptor GPR56/ADGRG1 on platelets. GPR56 is an adhesion G protein-coupled receptor that becomes activated following forced dissociation of its N-terminal fragment and C-terminal fragment or seven-transmembrane spanning domain (7TM). Fragment dissociation reveals the cryptic stalk of the 7TM, which acts as a tethered peptide agonist, and for GPR56, this activates platelet G13 signaling. GPR56 pharmacological probes activated platelets to undergo shape change and aggregation, which are critical for the formation of hemostatic plugs. Gpr56−/− mice exhibit prolonged bleeding, defective platelet plug formation in vessel injury assays, and delayed thrombotic vessel occlusion. Shear-force dependency of platelet adhesion to immobilized collagen was found to be GPR56 dependent. Circulating platelets roll along exposed collagen at vessel injury sites and respond with filipodia protrusion, shape change, and surface area expansion to facilitate platelet adhesion and plug formation. Various glycoproteins were considered to be both collagen responders and mediators of platelet adhesion, yet the signaling kinetics emanating from these receptors do not fully account for the rapid platelet cytoskeletal changes that occur in blood flow. We found the free N-terminal fragment of the adhesion G protein-coupled receptor (GPCR) GPR56 in human plasma and report that GPR56 is the platelet receptor that transduces signals from collagen and blood flow-induced shear force to activate G protein 13 signaling for platelet shape change. Gpr56−/− mice have prolonged bleeding, defective platelet plug formation, and delayed thrombotic occlusion. Human and mouse blood perfusion studies demonstrated GPR56 and shear-force dependence of platelet adhesion to immobilized collagen. Our work places GPR56 as an initial collagen responder and shear-force transducer that is essential for platelet shape change during hemostasis.
Collapse
|