1
|
Tian Y, Zong Y, Pang Y, Zheng Z, Ma Y, Zhang C, Gao J. Platelets and diseases: signal transduction and advances in targeted therapy. Signal Transduct Target Ther 2025; 10:159. [PMID: 40374650 DOI: 10.1038/s41392-025-02198-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/18/2024] [Accepted: 02/24/2025] [Indexed: 05/17/2025] Open
Abstract
Platelets are essential anucleate blood cells that play pivotal roles in hemostasis, tissue repair, and immune modulation. Originating from megakaryocytes in the bone marrow, platelets are small in size but possess a highly specialized structure that enables them to execute a wide range of physiological functions. The platelet cytoplasm is enriched with functional proteins, organelles, and granules that facilitate their activation and participation in tissue repair processes. Platelet membranes are densely populated with a variety of receptors, which, upon activation, initiate complex intracellular signaling cascades. These signaling pathways govern platelet activation, aggregation, and the release of bioactive molecules, including growth factors, cytokines, and chemokines. Through these mechanisms, platelets are integral to critical physiological processes such as thrombosis, wound healing, and immune surveillance. However, dysregulated platelet function can contribute to pathological conditions, including cancer metastasis, atherosclerosis, and chronic inflammation. Due to their central involvement in both normal physiology and disease, platelets have become prominent targets for therapeutic intervention. Current treatments primarily aim to modulate platelet signaling to prevent thrombosis in cardiovascular diseases or to reduce excessive platelet aggregation in other pathological conditions. Antiplatelet therapies are widely employed in clinical practice to mitigate clot formation in high-risk patients. As platelet biology continues to evolve, emerging therapeutic strategies focus on refining platelet modulation to enhance clinical outcomes and prevent complications associated with platelet dysfunction. This review explores the structure, signaling pathways, biological functions, and therapeutic potential of platelets, highlighting their roles in both physiological and pathological contexts.
Collapse
Affiliation(s)
- Yuchen Tian
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Yidan Pang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhikai Zheng
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiyang Ma
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Zhang S, Sun C, Huang Q, Du J, Xia Y, Zhou K, Yang B, Dai K, Yan R. The role of protein kinase C and the glycoprotein Ibα cytoplasmic tail in anti-glycoprotein Ibα antibody-induced platelet apoptosis and thrombocytopenia. Thromb Res 2024; 244:109210. [PMID: 39541612 DOI: 10.1016/j.thromres.2024.109210] [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: 04/23/2024] [Revised: 10/08/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Immune thrombocytopenia (ITP) is an autoimmune disease characterized by low platelet counts. ITP patients with anti-platelet glycoprotein (GP) Ibα (a subunit of GPIb-IX-V complex) autoantibodies, which induce Fc-independent signaling and platelet clearance, are refractory to conventional treatment. Protein kinase C (PKC) is activated by the binding of the ligand von Willebrand factor (VWF) to GPIbα and regulates VWF-GPIbα-induced platelet activation. However, the role of PKC in anti-GPIbα antibody-induced thrombocytopenia remains unknown. MATERIALS AND METHODS The anti-GPIbα antibody-induced PKC activation and its underlying mechanisms were first detected by Western blot, and then the effects of PKC inhibitors, PKC knockout, or GPIbα C-terminal removal on anti-GPIbα antibody-induced platelet apoptosis, activation, aggregation, and clearance were investigated by flow cytometry, platelet aggregometry, and platelet posttransfusion, respectively. Meanwhile, platelet retention and co-localization with macrophages in the liver were detected by spinning disc intravital confocal microscopy. RESULTS Anti-GPIbα antibody-induced PKC activation depends on GPIbα clustering and phosphoinositide 3-kinase (PI3K) activation and results in Akt phosphorylation. Pharmacologic inhibition or genetic ablation of PKC suppresses anti-GPIbα antibody-induced platelet apoptosis and activation. Moreover, the GPIbα cytoplasmic tail is required for antibody-induced PKC activation, platelet apoptosis, and activation. Inhibition or ablation of PKC and deletion of the GPIbα cytoplasmic tail protect platelets from clearance in vivo. CONCLUSIONS Our study indicates the important role of PKC and the GPIbα cytoplasmic tail in anti-GPIbα antibody-mediated platelet signaling and clearance and suggests a novel therapeutic target for ITP and other thrombocytopenic diseases.
Collapse
Affiliation(s)
- Sai Zhang
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, The First Affiliated Hospital Collaborative Innovation Center of Hematology, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, National Clinical Research Center for Hematological Diseases, Suzhou, China
| | - Chenglin Sun
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, The First Affiliated Hospital Collaborative Innovation Center of Hematology, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, National Clinical Research Center for Hematological Diseases, Suzhou, China
| | - Qiuxia Huang
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, The First Affiliated Hospital Collaborative Innovation Center of Hematology, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, National Clinical Research Center for Hematological Diseases, Suzhou, China
| | - Jiahao Du
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, The First Affiliated Hospital Collaborative Innovation Center of Hematology, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, National Clinical Research Center for Hematological Diseases, Suzhou, China
| | - Yue Xia
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, The First Affiliated Hospital Collaborative Innovation Center of Hematology, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, National Clinical Research Center for Hematological Diseases, Suzhou, China
| | - Kangxi Zhou
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, The First Affiliated Hospital Collaborative Innovation Center of Hematology, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, National Clinical Research Center for Hematological Diseases, Suzhou, China
| | - Biao Yang
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, The First Affiliated Hospital Collaborative Innovation Center of Hematology, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, National Clinical Research Center for Hematological Diseases, Suzhou, China
| | - Kesheng Dai
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, The First Affiliated Hospital Collaborative Innovation Center of Hematology, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, National Clinical Research Center for Hematological Diseases, Suzhou, China.
| | - Rong Yan
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, The First Affiliated Hospital Collaborative Innovation Center of Hematology, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, National Clinical Research Center for Hematological Diseases, Suzhou, China.
| |
Collapse
|
3
|
Zou J, Zhang P, Solari FA, Schönichen C, Provenzale I, Mattheij NJA, Kuijpers MJE, Rauch JS, Swieringa F, Sickmann A, Zieger B, Jurk K, Heemskerk JWM. Suppressed ORAI1-STIM1-dependent Ca 2+ entry by protein kinase C isoforms regulating platelet procoagulant activity. J Biol Chem 2024; 300:107899. [PMID: 39424145 PMCID: PMC11742345 DOI: 10.1016/j.jbc.2024.107899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024] Open
Abstract
Agonist-induced rises in cytosolic Ca2+ control most platelet responses in thrombosis and hemostasis. In human platelets, we earlier demonstrated that the ORAI1-STIM1 pathway is a major component of extracellular Ca2+ entry, in particular when induced via the ITAM-linked collagen receptor, glycoprotein VI (GPVI). In the present article, using functionally defective platelets from patients with a loss-of-function mutation in ORAI1 or STIM1, we show that Ca2+ entry induced by the endoplasmic reticulum ATPase inhibitor, thapsigargin, fully relies on this pathway. We demonstrate that both the GPVI-induced and thapsigargin-induced Ca2+ entry are strongly suppressed by protein kinase C (PKC) activation while leaving intracellular Ca2+ mobilization unchanged. Comparing the effects of a PKC inhibitory panel pointed to redundant roles of beta and theta PKC isoforms in Ca2+-entry suppression. In contrast, tyrosine kinases positively regulated GPVI-induced Ca2+ entry and mobilization. Label-free and stable isotope phosphoproteome analysis of GPVI-stimulated platelets suggested a regulatory role of bridging integrator-2 (BIN2), known as an important mediator of the ORAI1-STIM1 pathway in mouse platelets. Identified were 25 to 45 regulated phospho-sites in BIN2 and 16 to 18 in STIM1. Five of these were characterized as direct substrates of the expressed PKC isoforms alpha, beta delta, and theta. Functional platelet testing indicated that the downregulation of Ca2+ entry by PKC resulted in suppressed phosphatidylserine exposure and plasmatic thrombin generation. Conclusively, our results indicate that in platelets multiple PKC isoforms constrain the store-regulated Ca2+ entry via ORAI1-BIN2-STIM1, and hence downregulate platelet-dependent coagulation.
Collapse
Affiliation(s)
- Jinmi Zou
- Synapse Research Institute Maastricht, Maastricht, The Netherlands; Department of Biochemistry, CARIM, 6200 MD Maastricht University, Maastricht, The Netherlands
| | - Pengyu Zhang
- Department of Biochemistry, CARIM, 6200 MD Maastricht University, Maastricht, The Netherlands; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V, Dortmund, Germany
| | - Fiorella A Solari
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V, Dortmund, Germany
| | - Claudia Schönichen
- Department of Biochemistry, CARIM, 6200 MD Maastricht University, Maastricht, The Netherlands; Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Isabella Provenzale
- Department of Biochemistry, CARIM, 6200 MD Maastricht University, Maastricht, The Netherlands
| | - Nadine J A Mattheij
- Department of Clinical Chemistry and Hematology, Maxima Medical Center Veldhoven, Veldhoven, The Netherlands
| | - Marijke J E Kuijpers
- Department of Biochemistry, CARIM, 6200 MD Maastricht University, Maastricht, The Netherlands
| | - Julia S Rauch
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V, Dortmund, Germany
| | - Frauke Swieringa
- Synapse Research Institute Maastricht, Maastricht, The Netherlands
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V, Dortmund, Germany; Medizinische Fakultät, Medizinische Proteom-Center, Ruhr-Universität Bochum, Bochum, Germany; Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Barbara Zieger
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Johan W M Heemskerk
- Synapse Research Institute Maastricht, Maastricht, The Netherlands; Department of Biochemistry, CARIM, 6200 MD Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
4
|
Graff RC, Haimowitz A, Aguilan JT, Levine A, Zhang J, Yuan W, Roose-Girma M, Seshagiri S, Porcelli SA, Gamble MJ, Sidoli S, Bresnick AR, Backer JM. Platelet PI3Kβ regulates breast cancer metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.10.612261. [PMID: 39314490 PMCID: PMC11419023 DOI: 10.1101/2024.09.10.612261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Platelets promote tumor metastasis by several mechanisms. Platelet-tumor cell interactions induce the release of platelet cytokines, chemokines, and other factors that promote tumor cell epithelial-mesenchymal transition and invasion, granulocyte recruitment to circulating tumor cells (CTCs), and adhesion of CTCs to the endothelium, assisting in their extravasation at metastatic sites. Previous studies have shown that platelet activation in the context of thrombus formation requires the Class IA PI 3-kinase PI3Kβ. We now define a role for platelet PI3Kβ in breast cancer metastasis. Platelet PI3Kβ is essential for platelet-stimulated tumor cell invasion through Matrigel. Consistent with this finding, in vitro platelet-tumor cell binding and tumor cell-stimulated platelet activation are reduced in platelets isolated from PI3Kβ mutant mice. RNAseq and proteomic analysis of human breast epithelial cells co-cultured with platelets revealed that platelet PI3Kβ regulates the expression of EMT and metastasis-associated genes in these cells. The EMT and metastasis-associated proteins PAI-1 and IL-8 were specifically downregulated in co-cultures with PI3Kβ mutant platelets. PI3Kβ mutant platelets are impaired in their ability to stimulate YAP and Smad2 signaling in tumor cells, two pathways regulating PAI-1 expression. Finally, we show that mice expressing mutant PI3Kβ show reduced spontaneous metastasis, and platelets isolated from these mice are less able to stimulate experimental metastasis in WT mice. Taken together, these data support a role for platelet PI3Kβ in promoting breast cancer metastasis and highlight platelet PI3Kβ as a potential therapeutic target. Significance We demonstrate that platelet PI3Kβ regulates metastasis, broadening the potential use of PI3Kβ-selective inhibitors as novel agents to treat metastasis.
Collapse
|
5
|
Zou J, Sun S, De Simone I, ten Cate H, de Groot PG, de Laat B, Roest M, Heemskerk JW, Swieringa F. Platelet Activation Pathways Controlling Reversible Integrin αIIbβ3 Activation. TH OPEN 2024; 8:e232-e242. [PMID: 38911141 PMCID: PMC11193594 DOI: 10.1055/s-0044-1786987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/12/2024] [Indexed: 06/25/2024] Open
Abstract
Background Agonist-induced platelet activation, with the integrin αIIbβ3 conformational change, is required for fibrinogen binding. This is considered reversible under specific conditions, allowing a second phase of platelet aggregation. The signaling pathways that differentiate between a permanent or transient activation state of platelets are poorly elucidated. Objective To explore platelet signaling mechanisms induced by the collagen receptor glycoprotein VI (GPVI) or by protease-activated receptors (PAR) for thrombin that regulate time-dependent αIIbβ3 activation. Methods Platelets were activated with collagen-related peptide (CRP, stimulating GPVI), thrombin receptor-activating peptides, or thrombin (stimulating PAR1 and/or 4). Integrin αIIbβ3 activation and P-selectin expression was assessed by two-color flow cytometry. Signaling pathway inhibitors were applied before or after agonist addition. Reversibility of platelet spreading was studied by microscopy. Results Platelet pretreatment with pharmacological inhibitors decreased GPVI- and PAR-induced integrin αIIbβ3 activation and P-selectin expression in the target order of protein kinase C (PKC) > glycogen synthase kinase 3 > β-arrestin > phosphatidylinositol-3-kinase. Posttreatment revealed secondary αIIbβ3 inactivation (not P-selectin expression), in the same order, but this reversibility was confined to CRP and PAR1 agonist. Combined inhibition of conventional and novel PKC isoforms was most effective for integrin closure. Pre- and posttreatment with ticagrelor, blocking the P2Y 12 adenosine diphosphate (ADP) receptor, enhanced αIIbβ3 inactivation. Spreading assays showed that PKC or P2Y 12 inhibition provoked a partial conversion from filopodia to a more discoid platelet shape. Conclusion PKC and autocrine ADP signaling contribute to persistent integrin αIIbβ3 activation in the order of PAR1/GPVI > PAR4 stimulation and hence to stabilized platelet aggregation. These findings are relevant for optimization of effective antiplatelet treatment.
Collapse
Affiliation(s)
- Jinmi Zou
- Platelet (patho)physiology, Synapse Research Institute, Maastricht, The Netherlands
- Department of Biochemistry and Internal Medicine, Maastricht University Medical Center + , Maastricht, The Netherlands
| | - Siyu Sun
- Platelet (patho)physiology, Synapse Research Institute, Maastricht, The Netherlands
- Department of Biochemistry and Internal Medicine, Maastricht University Medical Center + , Maastricht, The Netherlands
| | - Ilaria De Simone
- Platelet (patho)physiology, Synapse Research Institute, Maastricht, The Netherlands
| | - Hugo ten Cate
- Department of Biochemistry and Internal Medicine, Maastricht University Medical Center + , Maastricht, The Netherlands
| | - Philip G. de Groot
- Platelet (patho)physiology, Synapse Research Institute, Maastricht, The Netherlands
| | - Bas de Laat
- Platelet (patho)physiology, Synapse Research Institute, Maastricht, The Netherlands
| | - Mark Roest
- Platelet (patho)physiology, Synapse Research Institute, Maastricht, The Netherlands
| | - Johan W.M. Heemskerk
- Platelet (patho)physiology, Synapse Research Institute, Maastricht, The Netherlands
| | - Frauke Swieringa
- Platelet (patho)physiology, Synapse Research Institute, Maastricht, The Netherlands
| |
Collapse
|
6
|
Fernández DI, Troitiño S, Sobota V, Tullemans BME, Zou J, van den Hurk H, García Á, Honarnejad S, Kuijpers MJE, Heemskerk JWM. Ultra-high throughput-based screening for the discovery of antiplatelet drugs affecting receptor dependent calcium signaling dynamics. Sci Rep 2024; 14:6229. [PMID: 38486006 PMCID: PMC10940705 DOI: 10.1038/s41598-024-56799-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024] Open
Abstract
Distinct platelet activation patterns are elicited by the tyrosine kinase-linked collagen receptor glycoprotein VI (GPVI) and the G-protein coupled protease-activated receptors (PAR1/4) for thrombin. This is reflected in the different platelet Ca2+ responses induced by the GPVI agonist collagen-related peptide (CRP) and the PAR1/4 agonist thrombin. Using a 96 well-plate assay with human Calcium-6-loaded platelets and a panel of 22 pharmacological inhibitors, we assessed the cytosolic Ca2+ signaling domains of these receptors and developed an automated Ca2+ curve algorithm. The algorithm was used to evaluate an ultra-high throughput (UHT) based screening of 16,635 chemically diverse small molecules with orally active physicochemical properties for effects on platelets stimulated with CRP or thrombin. Stringent agonist-specific selection criteria resulted in the identification of 151 drug-like molecules, of which three hit compounds were further characterized. The dibenzyl formamide derivative ANO61 selectively modulated thrombin-induced Ca2+ responses, whereas the aromatic sulfonyl imidazole AF299 and the phenothiazine ethopropazine affected CRP-induced responses. Platelet functional assays confirmed selectivity of these hits. Ethopropazine retained its inhibitory potential in the presence of plasma, and suppressed collagen-dependent thrombus buildup at arterial shear rate. In conclusion, targeting of platelet Ca2+ signaling dynamics in a screening campaign has the potential of identifying novel platelet-inhibiting molecules.
Collapse
Affiliation(s)
- Delia I Fernández
- The Department of Biochemistry, CARIM, Maastricht University, 6229 ER, Maastricht, The Netherlands
- Platelet Proteomics Group, CiMUS, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Sara Troitiño
- Platelet Proteomics Group, CiMUS, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Vladimír Sobota
- IHU-LIRYC, Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, 33604, Bordeaux, France
- Institut de Mathématiques de Bordeaux, UMR5251, University of Bordeaux, 33 405, Talence, France
| | - Bibian M E Tullemans
- The Department of Biochemistry, CARIM, Maastricht University, 6229 ER, Maastricht, The Netherlands
- Synapse Research Institute, Kon. Emmaplein 7, 6217 KD, Maastricht, The Netherlands
| | - Jinmi Zou
- The Department of Biochemistry, CARIM, Maastricht University, 6229 ER, Maastricht, The Netherlands
- Synapse Research Institute, Kon. Emmaplein 7, 6217 KD, Maastricht, The Netherlands
| | | | - Ángel García
- Platelet Proteomics Group, CiMUS, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | | | - Marijke J E Kuijpers
- The Department of Biochemistry, CARIM, Maastricht University, 6229 ER, Maastricht, The Netherlands.
- Thrombosis Expertise Centre, Heart and Vascular Centre, Maastricht University Medical Centre+, 6229 HX, Maastricht, The Netherlands.
| | - Johan W M Heemskerk
- The Department of Biochemistry, CARIM, Maastricht University, 6229 ER, Maastricht, The Netherlands.
- Synapse Research Institute, Kon. Emmaplein 7, 6217 KD, Maastricht, The Netherlands.
| |
Collapse
|
7
|
Gołaszewska A, Misztal T, Kazberuk A, Rusak T. Study on the Mechanism of the Adrenaline-Evoked Procoagulant Response in Human Platelets. Int J Mol Sci 2024; 25:2997. [PMID: 38474244 PMCID: PMC10932417 DOI: 10.3390/ijms25052997] [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: 01/20/2024] [Revised: 02/23/2024] [Accepted: 03/03/2024] [Indexed: 03/14/2024] Open
Abstract
Adrenaline has recently been found to trigger phosphatidylserine (PS) exposure on blood platelets, resulting in amplification of the coagulation process, but the mechanism is only fragmentarily established. Using a panel of platelet receptors' antagonists and modulators of signaling pathways, we evaluated the importance of these in adrenaline-evoked PS exposure by flow cytometry. Calcium and sodium ion influx into platelet cytosol, after adrenaline treatment, was examined by fluorimetric measurements. We found a strong reduction in PS exposure after blocking of sodium and calcium ion influx via Na+/H+ exchanger (NHE) and Na+/Ca2+ exchanger (NCX), respectively. ADP receptor antagonists produced a moderate inhibitory effect. Substantial limitation of PS exposure was observed in the presence of GPIIb/IIIa antagonist, phosphoinositide-3 kinase (PI3-K) inhibitors, or prostaglandin E1, a cyclic adenosine monophosphate (cAMP)-elevating agent. We demonstrated that adrenaline may develop a procoagulant response in human platelets with the substantial role of ion exchangers (NHE and NCX), secreted ADP, GPIIb/IIIa-dependent outside-in signaling, and PI3-K. Inhibition of the above mechanisms and increasing cytosolic cAMP seem to be the most efficient procedures to control adrenaline-evoked PS exposure in human platelets.
Collapse
Affiliation(s)
- Agata Gołaszewska
- Department of General and Experimental Pathology, Medical University of Bialystok, Mickiewicza 2C, 15-230 Bialystok, Poland
| | - Tomasz Misztal
- Department of Physical Chemistry, Medical University of Bialystok, Mickiewicza 2A, 15-369 Bialystok, Poland; (T.M.); (T.R.)
| | - Adam Kazberuk
- Department of Medicinal Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-959 Bialystok, Poland;
| | - Tomasz Rusak
- Department of Physical Chemistry, Medical University of Bialystok, Mickiewicza 2A, 15-369 Bialystok, Poland; (T.M.); (T.R.)
| |
Collapse
|
8
|
Guan IA, Liu JST, Sawyer RC, Li X, Jiao W, Jiramongkol Y, White MD, Hagimola L, Passam FH, Tran DP, Liu X, Schoenwaelder SM, Jackson SP, Payne RJ, Liu X. Integrating Phenotypic and Chemoproteomic Approaches to Identify Covalent Targets of Dietary Electrophiles in Platelets. ACS CENTRAL SCIENCE 2024; 10:344-357. [PMID: 38435523 PMCID: PMC10906253 DOI: 10.1021/acscentsci.3c00822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 12/24/2023] [Accepted: 12/28/2023] [Indexed: 03/05/2024]
Abstract
A large variety of dietary phytochemicals has been shown to improve thrombosis and stroke outcomes in preclinical studies. Many of these compounds feature electrophilic functionalities that potentially undergo covalent addition to the sulfhydryl side chain of cysteine residues within proteins. However, the impact of such covalent modifications on the platelet activity and function remains unclear. This study explores the irreversible engagement of 23 electrophilic phytochemicals with platelets, unveiling the unique antiplatelet selectivity of sulforaphane (SFN). SFN impairs platelet responses to adenosine diphosphate (ADP) and a thromboxane A2 receptor agonist while not affecting thrombin and collagen-related peptide activation. It also substantially reduces platelet thrombus formation under arterial flow conditions. Using an alkyne-integrated probe, protein disulfide isomerase A6 (PDIA6) was identified as a rapid kinetic responder to SFN. Mechanistic profiling studies revealed SFN's nuanced modulation of PDIA6 activity and substrate specificity. In an electrolytic injury model of thrombosis, SFN enhanced the thrombolytic activity of recombinant tissue plasminogen activator (rtPA) without increasing blood loss. Our results serve as a catalyst for further investigations into the preventive and therapeutic mechanisms of dietary antiplatelets, aiming to enhance the clot-busting power of rtPA, currently the only approved therapeutic for stroke recanalization that has significant limitations.
Collapse
Affiliation(s)
- Ivy A. Guan
- School
of Chemistry, Faculty of Science, The University
of Sydney, Sydney, New South Wales 2006, Australia
- The
Heart Research Institute, The University
of Sydney, Newtown, New South Wales 2042, Australia
| | - Joanna S. T. Liu
- The
Heart Research Institute, The University
of Sydney, Newtown, New South Wales 2042, Australia
- School
of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Renata C. Sawyer
- School
of Chemistry, Faculty of Science, The University
of Sydney, Sydney, New South Wales 2006, Australia
- The
Heart Research Institute, The University
of Sydney, Newtown, New South Wales 2042, Australia
| | - Xiang Li
- Department
of Medicine, Washington University in St.
Louis, St. Louis, Missouri 63110, United States
- McDonnell
Genome Institute, Washington University
in St. Louis, St. Louis, Missouri 63108, United States
| | - Wanting Jiao
- Ferrier Research
Institute, Victoria University of Wellington, Wellington 6140, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland 1142, New Zealand
| | - Yannasittha Jiramongkol
- School
of Chemistry, Faculty of Science, The University
of Sydney, Sydney, New South Wales 2006, Australia
- Charles
Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Mark D. White
- School
of Chemistry, Faculty of Science, The University
of Sydney, Sydney, New South Wales 2006, Australia
| | - Lejla Hagimola
- School
of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Freda H. Passam
- School
of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Denise P. Tran
- Sydney
Mass Spectrometry, The University of Sydney, Camperdown, New South Wales 2006, Australia
| | - Xiaoming Liu
- School
of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Simone M. Schoenwaelder
- The
Heart Research Institute, The University
of Sydney, Newtown, New South Wales 2042, Australia
- School
of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Shaun P. Jackson
- The
Heart Research Institute, The University
of Sydney, Newtown, New South Wales 2042, Australia
- Charles
Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Richard J. Payne
- School
of Chemistry, Faculty of Science, The University
of Sydney, Sydney, New South Wales 2006, Australia
- Australian
Research Council Centre of Excellence for Innovations in Peptide and
Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Xuyu Liu
- School
of Chemistry, Faculty of Science, The University
of Sydney, Sydney, New South Wales 2006, Australia
- The
Heart Research Institute, The University
of Sydney, Newtown, New South Wales 2042, Australia
| |
Collapse
|
9
|
Jooss NJ, Diender MG, Fernández DI, Huang J, Heubel-Moenen FCJ, van der Veer A, Kuijpers MJE, Poulter NS, Henskens YMC, Te Loo M, Heemskerk JWM. Restraining of glycoprotein VI- and integrin α2β1-dependent thrombus formation by platelet PECAM1. Cell Mol Life Sci 2024; 81:44. [PMID: 38236412 PMCID: PMC10796532 DOI: 10.1007/s00018-023-05058-2] [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: 07/14/2023] [Revised: 10/15/2023] [Accepted: 11/21/2023] [Indexed: 01/19/2024]
Abstract
The platelet receptors, glycoprotein VI (GPVI) and integrin α2β1 jointly control collagen-dependent thrombus formation via protein tyrosine kinases. It is unresolved to which extent the ITIM (immunoreceptor tyrosine-based inhibitory motif) receptor PECAM1 and its downstream acting protein tyrosine phosphatase PTPN11 interfere in this process. Here, we hypothesized that integrin α2β1 has a co-regulatory role in the PECAM1- and PTPN11-dependent restraint of thrombus formation. We investigated platelet activation under flow on collagens with a different GPVI dependency and using integrin α2β1 blockage. Blood was obtained from healthy subjects and from patients with Noonan syndrome with a gain-of-function mutation of PTPN11 and variable bleeding phenotype. On collagens with decreasing GPVI activity (types I, III, IV), the surface-dependent inhibition of PECAM1 did not alter thrombus parameters using control blood. Blockage of α2β1 generally reduced thrombus parameters, most effectively on collagen IV. Strikingly, simultaneous inhibition of PECAM1 and α2β1 led to a restoration of thrombus formation, indicating that the suppressing signaling effect of PECAM1 is masked by the platelet-adhesive receptor α2β1. Blood from 4 out of 6 Noonan patients showed subnormal thrombus formation on collagen IV. In these patients, effects of α2β1 blockage were counterbalanced by PECAM1 inhibition to a normal phenotype. In summary, we conclude that the suppression of GPVI-dependent thrombus formation by either PECAM1 or a gain-of-function of PTPN11 can be overruled by α2β1 engagement.
Collapse
Affiliation(s)
- Natalie J Jooss
- Department of Biochemistry, Maastricht University, Maastricht, The Netherlands
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Molecular Haematology Unit, University of Oxford, Headington, OX3 9DS, UK
| | - Marije G Diender
- Department of Pediatric Hematology, Amalia Children's Hospital, Radboud UMC, Nijmegen, The Netherlands
| | - Delia I Fernández
- Department of Biochemistry, Maastricht University, Maastricht, The Netherlands
- Platelet Proteomics Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Jingnan Huang
- Department of Biochemistry, Maastricht University, Maastricht, The Netherlands
- Platelet Proteomics Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Floor C J Heubel-Moenen
- Department of Internal Medicine, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Arian van der Veer
- Department of Pediatric Hematology, Amalia Children's Hospital, Radboud UMC, Nijmegen, The Netherlands
- Department of Pediatric Hematology, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | - Natalie S Poulter
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Nottingham, Midlands, UK
| | - Yvonne M C Henskens
- Central Diagnostic Laboratory, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Maroeska Te Loo
- Department of Pediatric Hematology, Amalia Children's Hospital, Radboud UMC, Nijmegen, The Netherlands
| | - Johan W M Heemskerk
- Department of Biochemistry, Maastricht University, Maastricht, The Netherlands.
- Synapse Research Institute Maastricht, Kon. Emmaplein 7, 6217 KD, Maastricht, The Netherlands.
| |
Collapse
|
10
|
Severin S, Gratacap MP, Bouvet L, Borret M, Kpotor AO, Chicanne G, Xuereb JM, Viaud J, Payrastre B. Phosphoinositides take a central stage in regulating blood platelet production and function. Adv Biol Regul 2024; 91:100992. [PMID: 37793962 DOI: 10.1016/j.jbior.2023.100992] [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: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/06/2023]
Abstract
Blood platelets are produced by megakaryocytes through a complex program of differentiation and play a critical role in hemostasis and thrombosis. These anucleate cells are the target of antithrombotic drugs that prevent them from clumping in cardiovascular disease conditions. Platelets also significantly contribute to various aspects of physiopathology, including interorgan communications, healing, inflammation, and thromboinflammation. Their production and activation are strictly regulated by highly elaborated mechanisms. Among them, those involving inositol lipids have drawn the attention of researchers. Phosphoinositides represent the seven combinatorially phosphorylated forms of the inositol head group of inositol lipids. They play a crucial role in regulating intracellular mechanisms, such as signal transduction, actin cytoskeleton rearrangements, and membrane trafficking, either by generating second messengers or by directly binding to specific domains of effector proteins. In this review, we will explore how phosphoinositides are implicated in controlling platelet production by megakaryocytes and in platelet activation processes. We will also discuss the diversity of phosphoinositides in platelets, their role in granule biogenesis and maintenance, as well as in integrin signaling. Finally, we will address the discovery of a novel pool of phosphatidylinositol 3-monophosphate in the outerleaflet of the plasma membrane of human and mouse platelets.
Collapse
Affiliation(s)
- Sonia Severin
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM UMR-1297 and Université Paul Sabatier, F-31432, Toulouse, France
| | - Marie-Pierre Gratacap
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM UMR-1297 and Université Paul Sabatier, F-31432, Toulouse, France
| | - Laura Bouvet
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM UMR-1297 and Université Paul Sabatier, F-31432, Toulouse, France
| | - Maxime Borret
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM UMR-1297 and Université Paul Sabatier, F-31432, Toulouse, France
| | - Afi Oportune Kpotor
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM UMR-1297 and Université Paul Sabatier, F-31432, Toulouse, France
| | - Gaëtan Chicanne
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM UMR-1297 and Université Paul Sabatier, F-31432, Toulouse, France
| | - Jean-Marie Xuereb
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM UMR-1297 and Université Paul Sabatier, F-31432, Toulouse, France
| | - Julien Viaud
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM UMR-1297 and Université Paul Sabatier, F-31432, Toulouse, France
| | - Bernard Payrastre
- Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), INSERM UMR-1297 and Université Paul Sabatier, F-31432, Toulouse, France; Laboratoire d'Hématologie, Centre de Référence des Pathologies Plaquettaires, Centre Hospitalier Universitaire de Toulouse Rangueil, F-31432, Toulouse, France.
| |
Collapse
|
11
|
Kim YJ, Kim TI, Lee A, Kim K, Hwang YH. Sinomenium acutum Modulates Platelet Aggregation and Thrombus Formation by Regulating the Glycoprotein VI-Mediated Signalosome in Mice. Pharmaceuticals (Basel) 2023; 17:6. [PMID: 38275992 PMCID: PMC10819422 DOI: 10.3390/ph17010006] [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: 11/06/2023] [Revised: 12/12/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
Sinomenium acutum (SA) has long been used as a traditional medicine in China, Japan, and Korea to treat a wide range of diseases. It has been traditionally used to ameliorate inflammation and improve blood circulation. However, its role in platelet activation has not been thoroughly investigated. Hence, we conducted this study to assess the potential inhibitory effect of SA on platelet aggregation and thrombus formation. The antiplatelet activities of SA were evaluated by assessing platelet aggregation, granular secretion, intracellular Ca2+ mobilization, and the Glycoprotein (GP) VI-mediated signalosome. The thrombosis and bleeding time assays were used to investigate the effect of SA (orally administered at 50 and 100 mg/kg for seven days) in mice. SA treatment at concentrations of 50, 100, and 200 μg/mL significantly reduced GPVI-mediated platelet aggregation, granular secretion, and intracellular Ca2+ mobilization. Further biochemical studies revealed that SA inhibited spleen tyrosine kinase, phospholipase Cγ2, phosphatidylinositol 3-kinase, and AKT phosphorylation. Interestingly, oral administration of SA efficiently ameliorated FeCl3-induced arterial thrombus formation without prolonging the tail bleeding time. These findings suggest that SA has beneficial effects in thrombosis and hemostasis. Therefore, SA holds promise as an effective therapeutic agent for the treatment of thrombotic diseases.
Collapse
Affiliation(s)
- Yeon-Ji Kim
- Korean Medicine-Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea
| | - Tae In Kim
- Korean Medicine-Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea
| | - Ami Lee
- Herbal Medicine Research Division, Korea Institution of Oriental Medicine, Daejeon 34054, Republic of Korea
- Korean Convergence Medical Science Major, KIOM School, University of Science & Technology (UST), Daejeon 34054, Republic of Korea
| | - Kyungho Kim
- Korean Medicine-Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea
- Korean Convergence Medical Science Major, KIOM School, University of Science & Technology (UST), Daejeon 34054, Republic of Korea
| | - Youn-Hwan Hwang
- Herbal Medicine Research Division, Korea Institution of Oriental Medicine, Daejeon 34054, Republic of Korea
- Korean Convergence Medical Science Major, KIOM School, University of Science & Technology (UST), Daejeon 34054, Republic of Korea
| |
Collapse
|
12
|
Gawaz M, Geisler T, Borst O. Current concepts and novel targets for antiplatelet therapy. Nat Rev Cardiol 2023; 20:583-599. [PMID: 37016032 DOI: 10.1038/s41569-023-00854-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/06/2023]
Abstract
Platelets have a crucial role in haemostasis and atherothrombosis. Pharmacological control of platelet hyper-reactivity has become a cornerstone in the prevention of thrombo-ischaemic complications in atherosclerotic diseases. Current antiplatelet therapies substantially improve clinical outcomes in patients with coronary artery disease, but at the cost of increased risk of bleeding. Beyond their role in thrombosis, platelets are known to regulate inflammatory (thrombo-inflammatory) and microcirculatory pathways. Therefore, controlling platelet hyper-reactivity might have implications for both tissue inflammation (myocardial ischaemia) and vascular inflammation (vulnerable plaque formation) to prevent atherosclerosis. In this Review, we summarize the pathophysiological role of platelets in acute myocardial ischaemia, vascular inflammation and atherosclerotic progression. Furthermore, we highlight current clinical concepts of antiplatelet therapy that have contributed to improving patient care and have facilitated more individualized therapy. Finally, we discuss novel therapeutic targets and compounds for antiplatelet therapy that are currently in preclinical development, some of which have a more favourable safety profile than currently approved drugs with regard to bleeding risk. These novel antiplatelet targets might offer new strategies to treat cardiovascular disease.
Collapse
Affiliation(s)
- Meinrad Gawaz
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany.
| | - Tobias Geisler
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
13
|
Schönichen C, Montague SJ, Brouns SL, Burston JJ, Cosemans JM, Jurk K, Kehrel BE, Koenen RR, Ní Áinle F, O’Donnell VB, Soehnlein O, Watson SP, Kuijpers MJ, Heemskerk JW, Nagy M. Antagonistic Roles of Human Platelet Integrin αIIbβ3 and Chemokines in Regulating Neutrophil Activation and Fate on Arterial Thrombi Under Flow. Arterioscler Thromb Vasc Biol 2023; 43:1700-1712. [PMID: 37409530 PMCID: PMC10443630 DOI: 10.1161/atvbaha.122.318767] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 06/14/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Platelets and neutrophils are the first blood cells accumulating at sites of arterial thrombus formation, and both cell types contribute to the pathology of thrombotic events. We aimed to identify key interaction mechanisms between these cells using microfluidic approaches. METHODS Whole-blood perfusion was performed over a collagen surface at arterial shear rate. Platelet and leukocyte (in majority neutrophil) activation were microscopically visualized using fluorescent markers. The contributions of platelet-adhesive receptors (integrin, P-selectin, CD40L) and chemokines were studied by using inhibitors or antibodies and using blood from patients with GT (Glanzmann thrombasthenia) lacking platelet-expressed αIIbβ3. RESULTS We observed (1) an unknown role of activated platelet integrin αIIbß3 preventing leukocyte adhesion, which was overcome by short-term flow disturbance provoking massive adhesion; (2) that platelet-expressed CD40L controls the crawling pattern and thrombus fidelity of the cells on a thrombus; (3) that continued secretion of platelet substances promotes activation of identified neutrophils, as assessed by (fMLP [N-formylmethionyl-leucyl-phenylalanine, a potent chemotactic agent and leukocyte activator] induced) [Ca2+]i rises and antigen expression; (4) and that platelet-released chemokines activate the adhered cells in the order of CXCL7>CCL5>CXCL4. Furthermore, postsilencing of the platelets in a thrombus suppressed the leukocyte activation. However, the leukocytes on thrombi did no more than limitedly form neutrophil extracellular traps, unless stimulated with phorbol ester or lipopolysaccharide. CONCLUSIONS Together, these findings reveal a multifaceted regulation of adhesion and activation of neutrophils by platelets in a thrombus, with a balanced role of several platelet-adhesive receptors and a promoting role of platelet-released substances. This multivalent nature of neutrophil-thrombus interactions offers novel prospects for pharmacological intervention.
Collapse
Affiliation(s)
- Claudia Schönichen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.S., S.L.N.B., J.M.E.M.C., R.R.K., S.P.W., M.J.E.K., J.W.M.H., M.N.)
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany (C.S., K.J.)
| | - Samantha J. Montague
- Institute of Cardiovascular Sciences, The Medical School, University of Birmingham, United Kingdom (S.J.M., S.P.W.)
| | - Sanne L.N. Brouns
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.S., S.L.N.B., J.M.E.M.C., R.R.K., S.P.W., M.J.E.K., J.W.M.H., M.N.)
| | - James J. Burston
- Systems Immunity Research Institute, School of Medicine, Cardiff University, United Kingdom (J.J.B., V.B.O.)
| | - Judith M.E.M. Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.S., S.L.N.B., J.M.E.M.C., R.R.K., S.P.W., M.J.E.K., J.W.M.H., M.N.)
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University of Mainz, Germany (C.S., K.J.)
- Department of Anaesthesiology and Intensive Care, University Hospital Muenster, Germany (K.J., B.E.K.)
| | - Beate E. Kehrel
- Department of Anaesthesiology and Intensive Care, University Hospital Muenster, Germany (K.J., B.E.K.)
| | - Rory R. Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.S., S.L.N.B., J.M.E.M.C., R.R.K., S.P.W., M.J.E.K., J.W.M.H., M.N.)
| | - Fionnuala Ní Áinle
- School of Medicine, University College Dublin, Ireland (F.N.Á.)
- Department of Haematology, Mater Misericordiae University Hospital and Rotunda Hospital, Dublin, Ireland (F.N.Á.)
| | - Valerie B. O’Donnell
- Systems Immunity Research Institute, School of Medicine, Cardiff University, United Kingdom (J.J.B., V.B.O.)
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Germany (O.S.)
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, Westfälische Wilhelms Universität, Münster, Germany (O.S.)
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (O.S.)
| | - Steve P. Watson
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.S., S.L.N.B., J.M.E.M.C., R.R.K., S.P.W., M.J.E.K., J.W.M.H., M.N.)
- Institute of Cardiovascular Sciences, The Medical School, University of Birmingham, United Kingdom (S.J.M., S.P.W.)
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, the Midlands, United Kingdom (S.P.W.)
| | - Marijke J.E. Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.S., S.L.N.B., J.M.E.M.C., R.R.K., S.P.W., M.J.E.K., J.W.M.H., M.N.)
- Thrombosis Expertise Centre, Heart and Vascular Centre, Maastricht University Medical Centre, the Netherlands (M.J.E.K.)
| | - Johan W.M. Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.S., S.L.N.B., J.M.E.M.C., R.R.K., S.P.W., M.J.E.K., J.W.M.H., M.N.)
- Synapse Research Institute, Maastricht, the Netherlands (J.W.M.H.)
| | - Magdolna Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (C.S., S.L.N.B., J.M.E.M.C., R.R.K., S.P.W., M.J.E.K., J.W.M.H., M.N.)
| |
Collapse
|
14
|
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
|
15
|
Schrottmaier WC, Kral-Pointner JB, Salzmann M, Mussbacher M, Schmuckenschlager A, Pirabe A, Brunnthaler L, Kuttke M, Maier B, Heber S, Datler H, Ekici Y, Niederreiter B, Heber U, Blomgren B, Gorki AD, Söderberg-Nauclér C, Payrastre B, Gratacap MP, Knapp S, Schabbauer G, Assinger A. Platelet p110β mediates platelet-leukocyte interaction and curtails bacterial dissemination in pneumococcal pneumonia. Cell Rep 2022; 41:111614. [PMID: 36351402 DOI: 10.1016/j.celrep.2022.111614] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/15/2022] [Accepted: 10/14/2022] [Indexed: 11/09/2022] Open
Abstract
Phosphatidylinositol 3-kinase catalytic subunit p110β is involved in tumorigenesis and hemostasis. However, it remains unclear if p110β also regulates platelet-mediated immune responses, which could have important consequences for immune modulation during anti-cancer treatment with p110β inhibitors. Thus, we investigate how platelet p110β affects inflammation and infection. Using a mouse model of Streptococcus pneumoniae-induced pneumonia, we find that both platelet-specific p110β deficiency and pharmacologic inhibition of p110β with TGX-221 exacerbate disease pathogenesis by preventing platelet-monocyte and neutrophil interactions, diminishing their infiltration and enhancing bacterial dissemination. Platelet p110β mediates neutrophil phagocytosis of S. pneumoniae in vitro and curtails bacteremia in vivo. Genetic deficiency or inhibition of platelet p110β also impairs macrophage recruitment in an independent model of sterile peritonitis. Our results demonstrate that platelet p110β dysfunction exacerbates pulmonary infection by impeding leukocyte functions. Thereby, our findings provide important insights into the immunomodulatory potential of PI3K inhibitors in bacterial infection.
Collapse
Affiliation(s)
- Waltraud Cornelia Schrottmaier
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Julia Barbara Kral-Pointner
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Manuel Salzmann
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Marion Mussbacher
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria; Department of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, University of Graz, 8010 Graz, Austria
| | - Anna Schmuckenschlager
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Anita Pirabe
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Laura Brunnthaler
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Mario Kuttke
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Barbara Maier
- Department of Medicine I, Research Division of Infection Biology, Medical University Vienna, 1090 Vienna, Austria
| | - Stefan Heber
- Institute of Physiology, Centre for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Hannes Datler
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Yasemin Ekici
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Birgit Niederreiter
- Division of Rheumatology, Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Ulrike Heber
- Department of Pathology and Comprehensive Cancer Centre, Medical University of Vienna, 1090 Vienna, Austria
| | - Bo Blomgren
- Department of Clinical Sciences, Danderyd Hospital, Department of Oncology-Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Anna-Dorothea Gorki
- Department of Medicine I, Research Division of Infection Biology, Medical University Vienna, 1090 Vienna, Austria
| | - Cecilia Söderberg-Nauclér
- Department of Medicine, Solna, Centre for Molecular Medicine, Microbial Pathogenesis Unit, Karolinska University Hospital, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Bernard Payrastre
- INSERM UMR1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Toulouse III Paul Sabatier, 31024 Toulouse, France
| | - Marie-Pierre Gratacap
- INSERM UMR1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Toulouse III Paul Sabatier, 31024 Toulouse, France
| | - Sylvia Knapp
- Department of Medicine I, Research Division of Infection Biology, Medical University Vienna, 1090 Vienna, Austria
| | - Gernot Schabbauer
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria.
| | - Alice Assinger
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria.
| |
Collapse
|
16
|
Reversible Platelet Integrin αIIbβ3 Activation and Thrombus Instability. Int J Mol Sci 2022; 23:ijms232012512. [PMID: 36293367 PMCID: PMC9604507 DOI: 10.3390/ijms232012512] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/28/2022] Open
Abstract
Integrin αIIbβ3 activation is essential for platelet aggregation and, accordingly, for hemostasis and arterial thrombosis. The αIIbβ3 integrin is highly expressed on platelets and requires an activation step for binding to fibrinogen, fibrin or von Willebrand factor (VWF). A current model assumes that the process of integrin activation relies on actomyosin force-dependent molecular changes from a bent-closed and extended-closed to an extended-open conformation. In this paper we review the pathways that point to a functional reversibility of platelet αIIbβ3 activation and transient aggregation. Furthermore, we refer to mouse models indicating that genetic defects that lead to reversible platelet aggregation can also cause instable thrombus formation. We discuss the platelet agonists and signaling pathways that lead to a transient binding of ligands to integrin αIIbβ3. Our analysis points to the (autocrine) ADP P2Y1 and P2Y12 receptor signaling via phosphoinositide 3-kinases and Akt as principal pathways linked to reversible integrin activation. Downstream signaling events by protein kinase C, CalDAG-GEFI and Rap1b have not been linked to transient integrin activation. Insight into the functional reversibility of integrin activation pathways will help to better understand the effects of antiplatelet agents.
Collapse
|
17
|
De Simone I, Baaten CCFMJ, Jandrot-Perrus M, Gibbins JM, ten Cate H, Heemskerk JWM, Jones CI, van der Meijden PEJ. Coagulation Factor XIIIa and Activated Protein C Activate Platelets via GPVI and PAR1. Int J Mol Sci 2022; 23:ijms231810203. [PMID: 36142125 PMCID: PMC9499330 DOI: 10.3390/ijms231810203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/24/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Platelet and coagulation activation are highly reciprocal processes driven by multi-molecular interactions. Activated platelets secrete several coagulation factors and expose phosphatidylserine, which supports the activation of coagulation factor proteins. On the other hand, the coagulation cascade generates known ligands for platelet receptors, such as thrombin and fibrin. Coagulation factor (F)Xa, (F)XIIIa and activated protein C (APC) can also bind to platelets, but the functional consequences are unclear. Here, we investigated the effects of the activated (anti)coagulation factors on platelets, other than thrombin. Multicolor flow cytometry and aggregation experiments revealed that the ‘supernatant of (hirudin-treated) coagulated plasma’ (SCP) enhanced CRP-XL-induced platelet responses, i.e., integrin αIIbβ3 activation, P-selectin exposure and aggregate formation. We demonstrated that FXIIIa in combination with APC enhanced platelet activation in solution, and separately immobilized FXIIIa and APC resulted in platelet spreading. Platelet activation by FXIIIa was inhibited by molecular blockade of glycoprotein VI (GPVI) or Syk kinase. In contrast, platelet spreading on immobilized APC was inhibited by PAR1 blockade. Immobilized, but not soluble, FXIIIa and APC also enhanced in vitro adhesion and aggregation under flow. In conclusion, in coagulation, factors other than thrombin or fibrin can induce platelet activation via GPVI and PAR receptors.
Collapse
Affiliation(s)
- Ilaria De Simone
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands
- School of Biological Sciences, Institute for Metabolic and Cardiovascular Research, University of Reading, Reading RG6 6AS, UK
| | - Constance C. F. M. J. Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands
- Institute for Molecular Cardiovascular Research, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Martine Jandrot-Perrus
- UMR_S1148, Laboratory for Vascular Translational Science, INSERM, University Paris Cité, F-75018 Paris, France
| | - Jonathan M. Gibbins
- School of Biological Sciences, Institute for Metabolic and Cardiovascular Research, University of Reading, Reading RG6 6AS, UK
| | - Hugo ten Cate
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
| | - Johan W. M. Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands
- Synapse Research Institute, 6217 KD Maastricht, The Netherlands
| | - Chris I. Jones
- School of Biological Sciences, Institute for Metabolic and Cardiovascular Research, University of Reading, Reading RG6 6AS, UK
- Correspondence: (C.I.J.); (P.E.J.v.d.M.); Tel.: +44-(0)-118-378-7047 (C.I.J.); +31-43-388-1684 (P.E.J.v.d.M.)
| | - Paola E. J. van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- Correspondence: (C.I.J.); (P.E.J.v.d.M.); Tel.: +44-(0)-118-378-7047 (C.I.J.); +31-43-388-1684 (P.E.J.v.d.M.)
| |
Collapse
|
18
|
Modulation of Glycoprotein VI and Its Downstream Signaling Pathways as an Antiplatelet Target. Int J Mol Sci 2022; 23:ijms23179882. [PMID: 36077280 PMCID: PMC9456422 DOI: 10.3390/ijms23179882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 11/16/2022] Open
Abstract
Antiplatelet therapy aims to reduce the risk of thrombotic events while maintaining hemostasis. A promising current approach is the inhibition of platelet glycoprotein GPVI-mediated adhesion pathways; pathways that do not involve coagulation. GPVI is a signaling receptor integral for collagen-induced platelet activation and participates in the thrombus consolidation process, being a suitable target for thrombosis prevention. Considering this, the blocking or antibody-mediated depletion of GPVI is a promising antiplatelet therapy for the effective and safe treatment of thrombotic diseases without a significant risk of bleeding and impaired hemostatic plug formation. This review describes the current knowledge concerning pharmaceutical approaches to platelet GPVI modulation and its downstream signaling pathways in this context.
Collapse
|
19
|
Zhou H, Wang L, Liu S, Wang W. The role of phosphoinositide 3-kinases in immune-inflammatory responses: potential therapeutic targets for abdominal aortic aneurysm. Cell Cycle 2022; 21:2339-2364. [PMID: 35792922 DOI: 10.1080/15384101.2022.2094577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The pathogenesis of abdominal aortic aneurysm (AAA) includes inflammatory responses, matrix metalloproteinases (MMPs) degradation, VSMC apoptosis, oxidative stress, and angiogenesis, among which the inflammatory response plays a key role. At present, surgery is the only curing treatment, and no effective drug can delay AAA progression in clinical practice. Therefore, searching for a signaling pathway related to the immune-inflammatory response is an essential direction for developing drugs targeting AAA. Recent studies have confirmed that the PI3K family plays an important role in many inflammatory diseases and is involved in regulating various cellular functions, especially in the immune-inflammatory response. This review focuses on the role of each isoform of PI3K in each stage of AAA immune-inflammatory response, making available explorations for a deeper understanding of the mechanism of inflammation and immune response during the formation and development of AAA.
Collapse
Affiliation(s)
- Haiyang Zhou
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Shuai Liu
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Wang
- Department of General &vascular Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
20
|
The Molecular Interaction of Collagen with Cell Receptors for Biological Function. Polymers (Basel) 2022; 14:polym14050876. [PMID: 35267698 PMCID: PMC8912536 DOI: 10.3390/polym14050876] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 01/25/2023] Open
Abstract
Collagen, an extracellular protein, covers the entire human body and has several important biological functions in normal physiology. Recently, collagen from non-human sources has attracted attention for therapeutic management and biomedical applications. In this regard, both land-based animals such as cow, pig, chicken, camel, and sheep, and marine-based resources such as fish, octopus, starfish, sea-cucumber, and jellyfish are widely used for collagen extraction. The extracted collagen is transformed into collagen peptides, hydrolysates, films, hydrogels, scaffolds, sponges and 3D matrix for food and biomedical applications. In addition, many strategic ideas are continuously emerging to develop innovative advanced collagen biomaterials. For this purpose, it is important to understand the fundamental perception of how collagen communicates with receptors of biological cells to trigger cell signaling pathways. Therefore, this review discloses the molecular interaction of collagen with cell receptor molecules to carry out cellular signaling in biological pathways. By understanding the actual mechanism, this review opens up several new concepts to carry out next level research in collagen biomaterials.
Collapse
|
21
|
Fernández DI, Provenzale I, Cheung HY, van Groningen J, Tullemans BM, Veninga A, Dunster JL, Honarnejad S, van den Hurk H, Kuijpers MJ, Heemskerk JW. Ultra-high-throughput Ca 2+ assay in platelets to distinguish ITAM-linked and G-protein-coupled receptor activation. iScience 2022; 25:103718. [PMID: 35072010 PMCID: PMC8762394 DOI: 10.1016/j.isci.2021.103718] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/12/2021] [Accepted: 12/29/2021] [Indexed: 12/30/2022] Open
Abstract
Antiplatelet drugs targeting G-protein-coupled receptors (GPCRs), used for the secondary prevention of arterial thrombosis, coincide with an increased bleeding risk. Targeting ITAM-linked receptors, such as the collagen receptor glycoprotein VI (GPVI), is expected to provide a better antithrombotic-hemostatic profile. Here, we developed and characterized an ultra-high-throughput (UHT) method based on intracellular [Ca2+]i increases to differentiate GPVI and GPCR effects on platelets. In 96-, 384-, or 1,536-well formats, Calcium-6-loaded human platelets displayed a slow-prolonged or fast-transient [Ca2+]i increase when stimulated with the GPVI agonist collagen-related peptide or with thrombin and other GPCR agonists, respectively. Semi-automated curve fitting revealed five parameters describing the Ca2+ responses. Verification of the UHT assay was done with a robustness compound library and clinically relevant platelet inhibitors. Taken together, these results present proof of principle of distinct receptor-type-dependent Ca2+ signaling curves in platelets, which allow identification of new inhibitors in a UHT way.
Collapse
Affiliation(s)
- Delia I. Fernández
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidad de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Isabella Provenzale
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
- Institute for Cardiovascular and Metabolic Research, University of Reading, RG6 6AX Reading, UK
| | - Hilaire Y.F. Cheung
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
- ISASLeibniz-Institut fur Analytische Wissenschaften-ISAS-e.V., 44227 Dortmund, Germany
- Institute of Cardiovascular Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | | | - Bibian M.E. Tullemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Alicia Veninga
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Joanne L. Dunster
- Institute for Cardiovascular and Metabolic Research, University of Reading, RG6 6AX Reading, UK
| | | | | | - Marijke J.E. Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
- Thrombosis Expertise Centre, Heart and Vascular Centre, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Johan W.M. Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
- Synapse Research Institute, Kon. Emmaplein 7, 6214 AC, Maastricht, the Netherlands
| |
Collapse
|
22
|
Schrottmaier WC, Mussbacher M, Salzmann M, Kral-Pointner JB, Assinger A. PI3K Isoform Signalling in Platelets. Curr Top Microbiol Immunol 2022; 436:255-285. [PMID: 36243848 DOI: 10.1007/978-3-031-06566-8_11] [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: 06/16/2023]
Abstract
Platelets are unique anucleated blood cells that constantly patrol the vasculature to seal and prevent injuries in a process termed haemostasis. Thereby they rapidly adhere to the subendothelial matrix and recruit further platelets, resulting in platelet aggregates. Apart from their central role in haemostasis, they also kept some of their features inherited by their evolutionary ancestor-the haemocyte, which was also involved in immune defences. Together with leukocytes, platelets fight pathogenic invaders and guide many immune processes. In addition, they rely on several signalling pathways which are also relevant to immune cells. Among these, one of the central signalling hubs is the PI3K pathway. Signalling processes in platelets are unique as they lack a nucleus and therefore transcriptional regulation is absent. As a result, PI3K subclasses fulfil distinct roles in platelets compared to other cells. In contrast to leukocytes, the central PI3K subclass in platelet signalling is PI3K class Iβ, which underlines the uniqueness of this cell type and opens new ways for potential platelet-specific pharmacologic inhibition. An overview of platelet function and signalling with emphasis on PI3K subclasses and their respective inhibitors is given in this chapter.
Collapse
Affiliation(s)
- Waltraud C Schrottmaier
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Marion Mussbacher
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, USA
| | - Manuel Salzmann
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Julia B Kral-Pointner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Alice Assinger
- Department of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
23
|
Navarro S, Stegner D, Nieswandt B, Heemskerk JWM, Kuijpers MJE. Temporal Roles of Platelet and Coagulation Pathways in Collagen- and Tissue Factor-Induced Thrombus Formation. Int J Mol Sci 2021; 23:ijms23010358. [PMID: 35008781 PMCID: PMC8745329 DOI: 10.3390/ijms23010358] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 12/31/2022] Open
Abstract
In hemostasis and thrombosis, the complex process of thrombus formation involves different molecular pathways of platelet and coagulation activation. These pathways are considered as operating together at the same time, but this has not been investigated. The objective of our study was to elucidate the time-dependency of key pathways of thrombus and clot formation, initiated by collagen and tissue factor surfaces, where coagulation is triggered via the extrinsic route. Therefore, we adapted a microfluidics whole-blood assay with the Maastricht flow chamber to acutely block molecular pathways by pharmacological intervention at desired time points. Application of the technique revealed crucial roles of glycoprotein VI (GPVI)-induced platelet signaling via Syk kinase as well as factor VIIa-induced thrombin generation, which were confined to the first minutes of thrombus buildup. A novel anti-GPVI Fab EMF-1 was used for this purpose. In addition, platelet activation with the protease-activating receptors 1/4 (PAR1/4) and integrin αIIbβ3 appeared to be prolongedly active and extended to later stages of thrombus and clot formation. This work thereby revealed a more persistent contribution of thrombin receptor-induced platelet activation than of collagen receptor-induced platelet activation to the thrombotic process.
Collapse
Affiliation(s)
- Stefano Navarro
- Institute of Experimental Biomedicine I, University Hospital Würzburg, Würzburg Josef-Schneider-Straße 2, 97080 Wurzburg, Germany; (S.N.); (D.S.); (B.N.)
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, 97080 Wurzburg, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - David Stegner
- Institute of Experimental Biomedicine I, University Hospital Würzburg, Würzburg Josef-Schneider-Straße 2, 97080 Wurzburg, Germany; (S.N.); (D.S.); (B.N.)
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, 97080 Wurzburg, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine I, University Hospital Würzburg, Würzburg Josef-Schneider-Straße 2, 97080 Wurzburg, Germany; (S.N.); (D.S.); (B.N.)
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, 97080 Wurzburg, Germany
| | - Johan W. M. Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
- Synapse Research Institute, Kon. Emmaplein 7, 6214 KD Maastricht, The Netherlands
- Correspondence: (J.W.M.H.); (M.J.E.K.); Tel.: +31-43-3881674 (M.J.E.K.)
| | - Marijke J. E. Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center+, Maastricht, Professor Debyelaan 25, 6229 HX Maastricht, The Netherlands
- Correspondence: (J.W.M.H.); (M.J.E.K.); Tel.: +31-43-3881674 (M.J.E.K.)
| |
Collapse
|
24
|
Veninga A, Baaten CCFMJ, De Simone I, Tullemans BME, Kuijpers MJE, Heemskerk JWM, van der Meijden PEJ. Effects of Platelet Agonists and Priming on the Formation of Platelet Populations. Thromb Haemost 2021; 122:726-738. [PMID: 34689320 PMCID: PMC9197595 DOI: 10.1055/s-0041-1735972] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Platelets from healthy donors display heterogeneity in responsiveness to agonists. The response thresholds of platelets are controlled by multiple bioactive molecules, acting as negatively or positively priming substances. Higher circulating levels of priming substances adenosine and succinate, as well as the occurrence of hypercoagulability, have been described for patients with ischaemic heart disease. Here, we present an improved methodology of flow cytometric analyses of platelet activation and the characterisation of platelet populations following activation and priming by automated clustering analysis.Platelets were treated with adenosine, succinate, or coagulated plasma before stimulation with CRP-XL, 2-MeSADP, or TRAP6 and labelled for activated integrin αIIbβ3 (PAC1), CD62P, TLT1, CD63, and GPIX. The Super-Enhanced Dmax subtraction algorithm and 2% marker (quadrant) setting were applied to identify populations, which were further defined by state-of-the-art clustering techniques (tSNE, FlowSOM).Following activation, five platelet populations were identified: resting, aggregating (PAC1 + ), secreting (α- and dense-granules; CD62P + , TLT1 + , CD63 + ), aggregating plus α-granule secreting (PAC1 + , CD62P + , TLT1 + ), and fully active platelet populations. The type of agonist determined the distribution of platelet populations. Adenosine in a dose-dependent way suppressed the fraction of fully activated platelets (TRAP6 > 2-MeSADP > CRP-XL), whereas succinate and coagulated plasma increased this fraction (CRP-XL > TRAP6 > 2-MeSADP). Interestingly, a subset of platelets showed a constant response (aggregating, secreting, or aggregating plus α-granule secreting), which was hardly affected by the stimulus strength or priming substances.
Collapse
Affiliation(s)
- Alicia Veninga
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| | - Constance C F M J Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands.,Institute for Molecular Cardiovascular Research, University Hospital Aachen, RWTH Aachen University, Germany
| | - Ilaria De Simone
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands.,Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, United Kingdom
| | - Bibian M E Tullemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| | - Marijke J E Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands.,Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| | - Paola E J van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands.,Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
25
|
Duan X, Perveen R, Dandamudi A, Adili R, Johnson J, Funk K, Berryman M, Davis AK, Holinstat M, Zheng Y, Akbar H. Pharmacologic targeting of Cdc42 GTPase by a small molecule Cdc42 activity-specific inhibitor prevents platelet activation and thrombosis. Sci Rep 2021; 11:13170. [PMID: 34162972 PMCID: PMC8222210 DOI: 10.1038/s41598-021-92654-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 05/27/2021] [Indexed: 01/14/2023] Open
Abstract
Gene targeting of Cdc42 GTPase has been shown to inhibit platelet activation. In this study, we investigated a hypothesis that inhibition of Cdc42 activity by CASIN, a small molecule Cdc42 Activity-Specific INhibitor, may down regulate platelet activation and thrombus formation. We investigated the effects of CASIN on platelet activation in vitro and thrombosis in vivo. In human platelets, CASIN, but not its inactive analog Pirl7, blocked collagen induced activation of Cdc42 and inhibited phosphorylation of its downstream effector, PAK1/2. Moreover, addition of CASIN to washed human platelets inhibited platelet spreading on immobilized fibrinogen. Treatment of human platelets with CASIN inhibited collagen or thrombin induced: (a) ATP secretion and platelet aggregation; and (b) phosphorylation of Akt, ERK and p38-MAPK. Pre-incubation of platelets with Pirl7, an inactive analog of CASIN, failed to inhibit collagen induced aggregation. Washing of human platelets after incubation with CASIN eliminated its inhibitory effect on collagen induced aggregation. Intraperitoneal administration of CASIN to wild type mice inhibited ex vivo aggregation induced by collagen but did not affect the murine tail bleeding times. CASIN administration, prior to laser-induced injury in murine cremaster muscle arterioles, resulted in formation of smaller and unstable thrombi compared to control mice without CASIN treatment. These data suggest that pharmacologic targeting of Cdc42 by specific and reversible inhibitors may lead to the discovery of novel antithrombotic agents.
Collapse
Affiliation(s)
- Xin Duan
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Rehana Perveen
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Akhila Dandamudi
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Reheman Adili
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - James Johnson
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Kevin Funk
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Mark Berryman
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Ashley Kuenzi Davis
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA.
| | - Huzoor Akbar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA.
| |
Collapse
|
26
|
Protein Disulphide Isomerase and NADPH Oxidase 1 Cooperate to Control Platelet Function and Are Associated with Cardiometabolic Disease Risk Factors. Antioxidants (Basel) 2021; 10:antiox10030497. [PMID: 33806982 PMCID: PMC8004975 DOI: 10.3390/antiox10030497] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/18/2021] [Accepted: 03/18/2021] [Indexed: 02/07/2023] Open
Abstract
Background: Protein disulphide isomerase (PDI) and NADPH oxidase 1 (Nox-1) regulate platelet function and reactive oxygen species (ROS) generation, suggesting potentially interdependent roles. Increased platelet reactivity and ROS production have been correlated with cardiometabolic disease risk factors. Objectives: To establish whether PDI and Nox-1 cooperate to control platelet function. Methods: Immunofluorescence microscopy was utilised to determine expression and localisation of PDI and Nox-1. Platelet aggregation, fibrinogen binding, P-selectin exposure, spreading and calcium mobilization were measured as markers of platelet function. A cross-sectional population study (n = 136) was conducted to assess the relationship between platelet PDI and Nox-1 levels and cardiometabolic risk factors. Results: PDI and Nox-1 co-localized upon activation induced by the collagen receptor GPVI. Co-inhibition of PDI and Nox-1 led to additive inhibition of GPVI-mediated platelet aggregation, activation and calcium flux. This was confirmed in murine Nox-1−/− platelets treated with PDI inhibitor bepristat, without affecting bleeding. PDI and Nox-1 together contributed to GPVI signalling that involved the phosphorylation of p38 MAPK, p47phox, PKC and Akt. Platelet PDI and Nox-1 levels were upregulated in obesity, with platelet Nox-1 also elevated in hypertensive individuals. Conclusions: We show that PDI and Nox-1 cooperate to control platelet function and are associated with cardiometabolic risk factors.
Collapse
|
27
|
Manjuprasanna VN, Urs AP, Rudresha GV, Milan Gowda MD, Jayachandra K, Hiremath V, Rajaiah R, Vishwanath BS. Drupin, a thrombin-like protease prompts platelet activation and aggregation through protease-activated receptors. J Cell Biochem 2021; 122:870-881. [PMID: 33748988 DOI: 10.1002/jcb.29917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/24/2021] [Accepted: 03/04/2021] [Indexed: 11/10/2022]
Abstract
Hemostasis is a proteolytically regulated process that requires activation of platelets and the blood coagulation cascade upon vascular injury. Activated platelets create a thrombogenic environment and amplify the coagulation process. Plant latex proteases (PLPs) have been used as therapeutic components to treat various ailments by folk healers. One of the main applications of plant latices is to stop bleeding from minor injuries and to enhance wound healing activity. Although many studies have reported the pro-coagulant activities of PLPs, an in-depth investigation is required to understand the mechanism of action of PLPs on platelets. Here, the effect of PLPs on platelet aggregation was studied systematically to validate the observed pharmacological effect by folk healers. Among 29 latices from the Ficus genus tested, Ficus drupacea exhibited potent pro-coagulant and thrombin-like activity. Drupin, a thrombin-like cysteine protease responsible for platelet aggregation was purified from F. drupacea latex. Drupin exhibits pro-coagulant activity and reduces the bleeding time in mice tail. It induces platelet aggregation by activating mitogen-activated protein kinases and the nuclear factor-κB and PI3K/Akt signalling cascade, which, in turn, phosphorylats, cytosolic phospholipase A2 leading to the release of thromboxane A2 from the granules to activate the nearby platelets to aggregate. Furthermore, we investigated the involvement of protease-activated receptors in drupin-induced platelet aggregation using specific protease activated receptor 1 (PAR1) and PAR4 receptor antagonists. The results confirmed that the drupin-induced platelet aggregation was mediated by both PAR1 and PAR4, synergistically. Overall, drupin reduces the bleeding time by exerting pro-coagulant activity and induces platelet aggregation by activating the intracellular signalling cascade.
Collapse
Affiliation(s)
| | - Amog P Urs
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| | - Gotravalli V Rudresha
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| | | | - Krishnegowda Jayachandra
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| | - Vilas Hiremath
- Vijayashree Diagnostics, Specialized Coagulation Lab, Bengaluru, Karnataka, India
| | - Rajesh Rajaiah
- Department of Studies in Molecular Biology, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| | - Bannikuppe S Vishwanath
- Department of Studies in Biochemistry, University of Mysore, Manasagangothri, Mysuru, Karnataka, India.,Department of Studies in Molecular Biology, University of Mysore, Manasagangothri, Mysuru, Karnataka, India
| |
Collapse
|
28
|
Phosphoinositide 3-kinases in platelets, thrombosis and therapeutics. Biochem J 2021; 477:4327-4342. [PMID: 33242335 DOI: 10.1042/bcj20190402] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/20/2020] [Accepted: 11/04/2020] [Indexed: 12/13/2022]
Abstract
Our knowledge on the expression, regulation and roles of the different phosphoinositide 3-kinases (PI3Ks) in platelet signaling and functions has greatly expanded these last twenty years. Much progress has been made in understanding the roles and regulations of class I PI3Ks which produce the lipid second messenger phosphatidylinositol 3,4,5 trisphosphate (PtdIns(3,4,5)P3). Selective pharmacological inhibitors and genetic approaches have allowed researchers to generate an impressive amount of data on the role of class I PI3Kα, β, δ and γ in platelet activation and in thrombosis. Furthermore, platelets do also express two class II PI3Ks (PI3KC2α and PI3KC2β), thought to generate PtdIns(3,4)P2 and PtdIns3P, and the sole class III PI3K (Vps34), known to synthesize PtdIns3P. Recent studies have started to reveal the importance of PI3KC2α and Vps34 in megakaryocytes and platelets, opening new perspective in our comprehension of platelet biology and thrombosis. In this review, we will summarize previous and recent advances on platelet PI3Ks isoforms. The implication of these kinases and their lipid products in fundamental platelet biological processes and thrombosis will be discussed. Finally, the relevance of developing potential antithrombotic strategies by targeting PI3Ks will be examined.
Collapse
|
29
|
van den Kerkhof DL, Nagy M, Wichapong K, Brouns SL, Heemskerk JWM, Hackeng TM, Dijkgraaf I. Inhibition of platelet adhesion, thrombus formation, and fibrin formation by a potent αIIbβ3 integrin inhibitor from ticks. Res Pract Thromb Haemost 2021; 5:231-242. [PMID: 33537548 PMCID: PMC7845065 DOI: 10.1002/rth2.12466] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 11/15/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Ticks puncture the skin of their hosts and secrete saliva, containing antiplatelet proteins, into the blood. Here, we studied disagregin, a potent platelet-inhibiting protein derived from the salivary glands of Ornithodoros moubata, an African soft tick. Whereas conventional αIIbβ3 antagonists contain an Arg-Gly-Asp (RGD) sequence for platelet integrin binding, disagregin contains an Arg-Glu-Asp (RED) sequence, hypothesizing a different mode of inhibitory action. OBJECTIVES We aimed to compare the inhibitory effects of disagregin and its RGD variant (RGD-disagregin) on platelet activation and to unravel the molecular basis of disagregin-αIIbβ3 integrin interactions. METHODS Disagregin and RGD-disagregin were synthesized by tert-butyloxycarbonyl -based solid-phase peptide synthesis. Effects of both disagregins on platelet aggregation were assessed by light transmission aggregometry in human platelet-rich plasma. Whole-blood thrombus formation was investigated by perfusing blood over collagen I with and without tissue factor at a high wall-shear rate (1000 s-1) in the presence of disagregin, RGD-disagregin, or eptifibatide. RESULTS Disagregin showed inhibition of collagen- and ADP-induced platelet aggregation with half maximal inhibitory concentration values of 64 and 99 nM, respectively. This resembled the complete antiaggregatory effect of eptifibatide. Multiparameter assessment of thrombus formation showed highly suppressed platelet adhesion and aggregate formation with both disagregins, in contrast to eptifibatide treatment, which incompletely blocked aggregation under flow. Fibrin formation under flow was delayed by both disagregin and RGD-disagregin (P < .01) and eptifibatide (P < .05). CONCLUSIONS Both αIIbβ3-blocking disagregins have a strong potential to suppress collagen-tissue factor-mediated platelet adhesion, thrombus formation, and fibrin formation. Both disagregins can be seen as potential new αIIbβ3 inhibitors.
Collapse
Affiliation(s)
- Danique L. van den Kerkhof
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Magdolna Nagy
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Kanin Wichapong
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Sanne L.N. Brouns
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Johan W. M. Heemskerk
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Tilman M. Hackeng
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Ingrid Dijkgraaf
- Department of BiochemistryCardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
30
|
Anderson R, Theron AJ, Steel HC, Nel JG, Tintinger GR. ADP-Mediated Upregulation of Expression of CD62P on Human Platelets Is Critically Dependent on Co-Activation of P2Y1 and P2Y12 Receptors. Pharmaceuticals (Basel) 2020; 13:ph13120420. [PMID: 33255391 PMCID: PMC7760858 DOI: 10.3390/ph13120420] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
This study probed the differential utilization of P2Y1 and P2Y12 receptors in mobilizing CD62P (P-selectin) from intracellular granules following activation of human platelets with adenosine 5′-diphosphate (ADP, 100 µmol·L−1) Platelet-rich plasma (PRP) was prepared from the blood of adult humans. CD62P was measured by flow cytometry following activation of PRP with ADP in the absence and presence of the selective antagonists of P2Y1 and P2Y12 receptors, MRS2500 and PSB0739 (both 0.155–10 µmol·L−1), respectively. Effects of the test agents on ADP-activated, CD62P-dependent formation of neutrophil:platelet (NP) aggregates were also measured by flow cytometry, while phosphatidylinositol 3-kinase (PI3K) activity was measured according to Akt1 phosphorylation in platelet lysates. Treatment with MRS2500 or PSB0739 at 10 µmol·L−1 almost completely attenuated (94.6% and 86% inhibition, respectively) ADP-activated expression of CD62P and also inhibited NP aggregate formation. To probe the mechanisms involved in P2Y1/P2Y12 receptor-mediated expression of CD62P, PRP was pre-treated with U73122 (phospholipase C (PLC) inhibitor), 2-aminoethoxy-diphenyl borate (2-APB, inositol triphosphate receptor antagonist), calmidazolium chloride (calmodulin inhibitor), or wortmannin (PI3K inhibitor). U73122, 2-APB, and wortmannin caused almost complete inhibition of ADP-activated expression of CD62P, while calmidazolium chloride caused statistically significant, partial inhibition. PSB0739, but not MRS2500, caused potent inhibition of PI3K-mediated phosphorylation of Akt1. Optimal mobilization of CD62P by ADP-stimulated platelets is critically dependent on the co-activation of platelet P2Y1 and P2Y12 receptors. P2Y12 receptor activation is the key event in activation of PI3K, while activation of the P2Y1 receptor appears to create a high cytosolic Ca2+ environment conducive to optimum PI3K activity.
Collapse
Affiliation(s)
- Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (A.J.T.); (H.C.S.)
- Correspondence: ; Tel.: +27-12-318-2425; Fax: +27-12-323-0732
| | - Annette J. Theron
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (A.J.T.); (H.C.S.)
| | - Helen C. Steel
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (A.J.T.); (H.C.S.)
| | - Jan G. Nel
- Department of Haematology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
- Tshwane Academic Division of the National Laboratory Health Service of South Africa, Pretoria 0001, South Africa
| | - Gregory R. Tintinger
- Department of Internal Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| |
Collapse
|
31
|
Impaired iloprost-induced platelet inhibition and phosphoproteome changes in patients with confirmed pseudohypoparathyroidism type Ia, linked to genetic mutations in GNAS. Sci Rep 2020; 10:11389. [PMID: 32647264 PMCID: PMC7347634 DOI: 10.1038/s41598-020-68379-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/22/2020] [Indexed: 11/16/2022] Open
Abstract
Patients diagnosed with pseudohypoparathyroidism type Ia (PHP Ia) suffer from hormonal resistance and abnormal postural features, in a condition classified as Albright hereditary osteodystrophy (AHO) syndrome. This syndrome is linked to a maternally inherited mutation in the GNAS complex locus, encoding for the GTPase subunit Gsα. Here, we investigated how platelet phenotype and omics analysis can assist in the often difficult diagnosis. By coupling to the IP receptor, Gsα induces platelet inhibition via adenylyl cyclase and cAMP-dependent protein kinase A (PKA). In platelets from seven patients with suspected AHO, one of the largest cohorts examined, we studied the PKA-induced phenotypic changes. Five patients with a confirmed GNAS mutation, displayed impairments in Gsα-dependent VASP phosphorylation, aggregation, and microfluidic thrombus formation. Analysis of the platelet phosphoproteome revealed 2,516 phosphorylation sites, of which 453 were regulated by Gsα-PKA. Common changes in the patients were: (1) a joint panel of upregulated and downregulated phosphopeptides; (2) overall PKA dependency of the upregulated phosphopeptides; (3) links to key platelet function pathways. In one patient with GNAS mutation, diagnosed as non-AHO, the changes in platelet phosphoproteome were reversed. This combined approach thus revealed multiple phenotypic and molecular biomarkers to assist in the diagnosis of suspected PHP Ia.
Collapse
|
32
|
|
33
|
Durrant TN, Hers I. PI3K inhibitors in thrombosis and cardiovascular disease. Clin Transl Med 2020; 9:8. [PMID: 32002690 PMCID: PMC6992830 DOI: 10.1186/s40169-020-0261-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/13/2020] [Indexed: 12/15/2022] Open
Abstract
Phosphoinositide 3-kinases (PI3Ks) are lipid kinases that regulate important intracellular signalling and vesicle trafficking events via the generation of 3-phosphoinositides. Comprising eight core isoforms across three classes, the PI3K family displays broad expression and function throughout mammalian tissues, and the (patho)physiological roles of these enzymes in the cardiovascular system present the PI3Ks as potential therapeutic targets in settings such as thrombosis, atherosclerosis and heart failure. This review will discuss the PI3K enzymes and their roles in cardiovascular physiology and disease, with a particular focus on platelet function and thrombosis. The current progress and future potential of targeting the PI3K enzymes for therapeutic benefit in cardiovascular disease will be considered, while the challenges of developing drugs against these master cellular regulators will be discussed.
Collapse
Affiliation(s)
- Tom N Durrant
- Department of Chemistry, University of Oxford, Oxford, OX1 3QZ, UK.
| | - Ingeborg Hers
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.
| |
Collapse
|
34
|
Abstract
Platelets - blood cells continuously produced from megakaryocytes mainly in the bone marrow - are implicated not only in haemostasis and arterial thrombosis, but also in other physiological and pathophysiological processes. This Review describes current evidence for the heterogeneity in platelet structure, age, and activation properties, with consequences for a diversity of platelet functions. Signalling processes of platelet populations involved in thrombus formation with ongoing coagulation are well understood. Genetic approaches have provided information on multiple genes related to normal haemostasis, such as those encoding receptors and signalling or secretory proteins, that determine platelet count and/or responsiveness. As highly responsive and secretory cells, platelets can alter the environment through the release of growth factors, chemokines, coagulant factors, RNA species, and extracellular vesicles. Conversely, platelets will also adapt to their environment. In disease states, platelets can be positively primed to reach a pre-activated condition. At the inflamed vessel wall, platelets interact with leukocytes and the coagulation system, interactions mediating thromboinflammation. With current antiplatelet therapies invariably causing bleeding as an undesired adverse effect, novel therapies can be more beneficial if directed against specific platelet responses, populations, interactions, or priming conditions. On the basis of these novel concepts and processes, we discuss several initiatives to target platelets therapeutically.
Collapse
|
35
|
Makhoul S, Trabold K, Gambaryan S, Tenzer S, Pillitteri D, Walter U, Jurk K. cAMP- and cGMP-elevating agents inhibit GPIbα-mediated aggregation but not GPIbα-stimulated Syk activation in human platelets. Cell Commun Signal 2019; 17:122. [PMID: 31519182 PMCID: PMC6743169 DOI: 10.1186/s12964-019-0428-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 08/29/2019] [Indexed: 12/29/2022] Open
Abstract
Background The glycoprotein (GP) Ib-IX-V complex is a unique platelet plasma membrane receptor, which is essential for platelet adhesion and thrombus formation. GPIbα, part of the GPIb-IX-V complex, has several physiological ligands such as von Willebrand factor (vWF), thrombospondin and distinct coagulation factors, which trigger platelet activation. Despite having an important role, intracellular GPIb-IX-V signaling and its regulation by other pathways are not well defined. Our aim was to establish the intracellular signaling response of selective GPIbα activation in human platelets, in particular the role of the tyrosine kinase Syk and its regulation by cAMP/PKA and cGMP/PKG pathways, respectively. We addressed this using echicetin beads (EB), which selectively bind to GPIbα and induce platelet aggregation. Methods Purified echicetin from snake Echis carinatus venom was validated by mass spectrometry. Washed human platelets were incubated with EB, in the presence or absence of echicetin monomers (EM), Src family kinase (SFK) inhibitors, Syk inhibitors and the cAMP- and cGMP-elevating agents iloprost and riociguat, respectively. Platelet aggregation was analyzed by light transmission aggregometry, protein phosphorylation by immunoblotting. Intracellular messengers inositolmonophosphate (InsP1) and Ca2+i were measured by ELISA and Fluo-3 AM/FACS, respectively. Results EB-induced platelet aggregation was dependent on integrin αIIbβ3 and secondary mediators ADP and TxA2, and was antagonized by EM. EB stimulated Syk tyrosine phosphorylation at Y352, which was SFK-dependent and Syk-independent, whereas Y525/526 phosphorylation was SFK-dependent and partially Syk-dependent. Furthermore, phosphorylation of both Syk Y352 and Y525/526 was completely integrin αIIbβ3-independent but, in the case of Y525/526, was partially ADP/TxA2-dependent. Syk activation, observed as Y352/ Y525/Y526 phosphorylation, led to the phosphorylation of direct substrates (LAT Y191, PLCγ2 Y759) and additional targets (Akt S473). PKA/PKG pathways inhibited EB-induced platelet aggregation and Akt phosphorylation but, surprisingly, enhanced Syk and LAT/PLCγ2 tyrosine phosphorylation. A similar PKA/PKG effect was confirmed with convulxin−/GPVI-stimulated platelets. EB-induced InsP1 accumulation/InsP3 production and Ca2+-release were Syk-dependent, but only partially inhibited by PKA/PKG pathways. Conclusion EB and EM are specific agonists and antagonists, respectively, of GPIbα-mediated Syk activation leading to platelet aggregation. The cAMP/PKA and cGMP/PKG pathways do not inhibit but enhance GPIbα−/GPVI-initiated, SFK-dependent Syk activation, but strongly inhibit further downstream responses including aggregation. These data establish an important intracellular regulatory network induced by GPIbα. Graphical abstract ![]()
Electronic supplementary material The online version of this article (10.1186/s12964-019-0428-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stephanie Makhoul
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Katharina Trabold
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stepan Gambaryan
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz of the Johannes Gutenberg University Mainz, Mainz, Germany.,Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
| | - Stefan Tenzer
- Core Facility for Mass Spectrometry, Institute for Immunology, University Medical Center Mainz, Mainz, Germany
| | | | - Ulrich Walter
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
36
|
Platelet heterogeneity in activation-induced glycoprotein shedding: functional effects. Blood Adv 2019; 2:2320-2331. [PMID: 30232085 DOI: 10.1182/bloodadvances.2017011544] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 08/15/2018] [Indexed: 12/20/2022] Open
Abstract
The platelet receptors glycoprotein Ibα (GPIbα) and GPVI are known to be cleaved by members of a disintegrin and metalloprotease (ADAM) family (ADAM10 and ADAM17), but the mechanisms and consequences of this shedding are not well understood. Our results revealed that (1) glycoprotein shedding is confined to distinct platelet populations showing near-complete shedding, (2) the heterogeneity between (non)shed platelets is independent of agonist type but coincides with exposure of phosphatidylserine (PS), and (3) distinct pathways of shedding are induced by elevated Ca2+, low Ca2+ protein kinase C (PKC), or apoptotic activation. Furthermore, we found that receptor shedding reduces binding of von Willebrand factor, enhances binding of coagulation factors, and augments fibrin formation. In response to Ca2+-increasing agents, shedding of GPIbα was abolished by ADAM10/17 inhibition but not by blockage of calpain. Stimulation of PKC induced shedding of only GPIbα, which was annulled by kinase inhibition. The proapoptotic agent ABT-737 induced shedding, which was caspase dependent. In Scott syndrome platelets that are deficient in Ca2+-dependent PS exposure, shedding occurred normally, indicating that PS exposure is not a prerequisite for ADAM activity. In whole-blood thrombus formation, ADAM-dependent glycoprotein shedding enhanced thrombin generation and fibrin formation. Together, these findings indicate that 2 major activation pathways can evoke ADAM-mediated glycoprotein shedding in distinct platelet populations and that shedding modulates platelet function from less adhesive to more procoagulant.
Collapse
|
37
|
Szydzik C, Brazilek RJ, Akbaridoust F, de Silva C, Moon M, Marusic I, Ooi ASH, Nandurkar HH, Hamilton JR, Mitchell A, Nesbitt WS. Active Micropump-Mixer for Rapid Antiplatelet Drug Screening in Whole Blood. Anal Chem 2019; 91:10830-10839. [PMID: 31343155 DOI: 10.1021/acs.analchem.9b02486] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
There is a need for scalable automated lab-on-chip systems incorporating precise hemodynamic control that can be applied to high-content screening of new more efficacious antiplatelet therapies. This paper reports on the development and characterization of a novel active micropump-mixer microfluidic to address this need. Using a novel reciprocating elastomeric micropump design, we take advantage of the flexible structural and actuation properties of this framework to manage the hemodynamics for on-chip platelet thrombosis assay on type 1 fibrillar collagen, using whole blood. By characterizing and harnessing the complex three-dimensional hemodynamics of the micropump operation in conjunction with a microvalve controlled reagent injection system we demonstrate that this prototype can act as a real-time assay of antiplatelet drug pharmacokinetics. In a proof-of-concept preclinical application, we utilize this system to investigate the way in which rapid dosing of human whole blood with isoform selective inhibitors of phosphatidylinositol 3-kinase dose dependently modulate platelet thrombus dynamics. This modular system exhibits utility as an automated multiplexable assay system with applications to high-content chemical library screening of new antiplatelet therapies.
Collapse
Affiliation(s)
- Crispin Szydzik
- The Australian Centre for Blood Diseases , Monash University , 99 Commercial Road , Melbourne , Victoria 3004 , Australia.,School of Engineering , RMIT University , 124 La Trobe Street , Melbourne , Victoria 3000 , Australia
| | - Rose J Brazilek
- The Australian Centre for Blood Diseases , Monash University , 99 Commercial Road , Melbourne , Victoria 3004 , Australia
| | - Farzan Akbaridoust
- School of Engineering , RMIT University , 124 La Trobe Street , Melbourne , Victoria 3000 , Australia.,Department of Mechanical Engineering, Melbourne School of Engineering , The University of Melbourne , Melbourne , Victoria 3010 , Australia
| | - Charitha de Silva
- Department of Mechanical Engineering, Melbourne School of Engineering , The University of Melbourne , Melbourne , Victoria 3010 , Australia.,School of Mechanical and Manufacturing Engineering , The University of New South Wales , Sydney , New South Wales 2052 , Australia
| | - Mitchell Moon
- The Australian Centre for Blood Diseases , Monash University , 99 Commercial Road , Melbourne , Victoria 3004 , Australia
| | - Ivan Marusic
- Department of Mechanical Engineering, Melbourne School of Engineering , The University of Melbourne , Melbourne , Victoria 3010 , Australia
| | - Andrew S H Ooi
- Department of Mechanical Engineering, Melbourne School of Engineering , The University of Melbourne , Melbourne , Victoria 3010 , Australia
| | - Harshal H Nandurkar
- The Australian Centre for Blood Diseases , Monash University , 99 Commercial Road , Melbourne , Victoria 3004 , Australia
| | - Justin R Hamilton
- The Australian Centre for Blood Diseases , Monash University , 99 Commercial Road , Melbourne , Victoria 3004 , Australia
| | - Arnan Mitchell
- School of Engineering , RMIT University , 124 La Trobe Street , Melbourne , Victoria 3000 , Australia
| | - Warwick S Nesbitt
- The Australian Centre for Blood Diseases , Monash University , 99 Commercial Road , Melbourne , Victoria 3004 , Australia.,School of Engineering , RMIT University , 124 La Trobe Street , Melbourne , Victoria 3000 , Australia
| |
Collapse
|
38
|
Bresnick AR, Backer JM. PI3Kβ-A Versatile Transducer for GPCR, RTK, and Small GTPase Signaling. Endocrinology 2019; 160:536-555. [PMID: 30601996 PMCID: PMC6375709 DOI: 10.1210/en.2018-00843] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/20/2018] [Indexed: 12/17/2022]
Abstract
The phosphoinositide 3-kinase (PI3K) family includes eight distinct catalytic subunits and seven regulatory subunits. Only two PI3Ks are directly regulated downstream from G protein-coupled receptors (GPCRs): the class I enzymes PI3Kβ and PI3Kγ. Both enzymes produce phosphatidylinositol 3,4,5-trisposphate in vivo and are regulated by both heterotrimeric G proteins and small GTPases from the Ras or Rho families. However, PI3Kβ is also regulated by direct interactions with receptor tyrosine kinases (RTKs) and their tyrosine phosphorylated substrates, and similar to the class II and III PI3Ks, it binds activated Rab5. The unusually complex regulation of PI3Kβ by small and trimeric G proteins and RTKs leads to a rich landscape of signaling responses at the cellular and organismic levels. This review focuses first on the regulation of PI3Kβ activity in vitro and in cells, and then summarizes the biology of PI3Kβ signaling in distinct tissues and in human disease.
Collapse
Affiliation(s)
- Anne R Bresnick
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
| | - Jonathan M Backer
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
39
|
|
40
|
Akbar H, Duan X, Piatt R, Saleem S, Davis AK, Tandon NN, Bergmeier W, Zheng Y. Small molecule targeting the Rac1-NOX2 interaction prevents collagen-related peptide and thrombin-induced reactive oxygen species generation and platelet activation. J Thromb Haemost 2018; 16:2083-2096. [PMID: 30007118 PMCID: PMC6472274 DOI: 10.1111/jth.14240] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Indexed: 12/29/2022]
Abstract
Essentials Reactive oxygen species (ROS) generation by NOX2 plays a critical role in platelet activation. Rac1 regulation of NOX2 is important for ROS generation. Small molecule inhibitor of the Rac1-p67phox interaction prevents platelet activation. Pharmacologic targeting of Rac1-NOX2 axis can be a viable approach for antithrombotic therapy. SUMMARY Background Platelets from patients with X-linked chronic granulomatous disease or mice deficient in nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) oxidase isoform NOX2 exhibit diminished reactive oxygen species (ROS) generation and platelet activation. Binding of Rac1 GTPase to p67phox plays a critical role in NOX2 activation by facilitating the assembly of the NOX2 enzyme complex. Objective We tested the hypothesis that Phox-I, a rationally designed small molecule inhibitor of Rac-p67phox interaction, may serve as an antithrombosis agent by suppressing ROS production and platelet activation. Results Collagen-related peptide (CRP) induced ROS generation in a time-dependent manner. Platelets from Rac1-/- mice or human platelets treated with NSC23766, a specific Rac inhibitor, produced significantly less ROS in response to CRP. Treatment of platelets with Phox-I inhibited diverse CRP-induced responses, including: (i) ROS generation; (ii) release of P-selectin; (iii) secretion of ATP; (iv) platelet aggregation; and (v) phosphorylation of Akt. Similarly, incubation of platelets with Phox-I inhibited thrombin-induced: (i) secretion of ATP; (ii) platelet aggregation; (iii) rise in cytosolic calcium; and (iv) phosphorylation of Akt. In mouse models, intraperitoneal administration of Phox-I inhibited: (i) collagen-induced platelet aggregation without affecting the tail bleeding time and (ii) in vivo platelet adhesion/accumulation at the laser injury sites on the saphenous vein without affecting the time for complete cessation of blood loss. Conclusions Small molecule targeting of the Rac1-p67phox interaction may present an antithrombosis regimen by preventing GPVI- and non-GPVI-mediated NOX2 activation, ROS generation and platelet function without affecting the bleeding time.
Collapse
Affiliation(s)
- H Akbar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - X Duan
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - R Piatt
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - S Saleem
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - A K Davis
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | | | - W Bergmeier
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Y Zheng
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
41
|
Laurent PA, Hechler B, Solinhac R, Ragab A, Cabou C, Anquetil T, Severin S, Denis CV, Mangin PH, Vanhaesebroeck B, Payrastre B, Gratacap MP. Impact of PI3Kα (Phosphoinositide 3-Kinase Alpha) Inhibition on Hemostasis and Thrombosis. Arterioscler Thromb Vasc Biol 2018; 38:2041-2053. [PMID: 30354258 DOI: 10.1161/atvbaha.118.311410] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective- PI3Kα (phosphoinositide 3-kinase alpha) is a therapeutic target in oncology, but its role in platelets and thrombosis remains ill characterized. In this study, we have analyzed the role of PI3Kα in vitro, ex vivo, and in vivo in 2 models of arterial thrombosis. Approach and Results- Using mice selectively deficient in p110α in the megakaryocyte lineage and isoform-selective inhibitors, we confirm that PI3Kα is not mandatory but participates to thrombus growth over a collagen matrix at arterial shear rate. Our data uncover a role for PI3Kα in low-level activation of the GP (glycoprotein) VI-collagen receptor by contributing to ADP secretion and in turn full activation of PI3Kβ and Akt/PKB (protein kinase B). This effect was no longer observed at high level of GP VI agonist concentration. Our study also reveals that over a vWF (von Willebrand factor) matrix, PI3Kα regulates platelet stationary adhesion contacts under arterial flow through its involvement in the outside-in signaling of vWF-engaged αIIbβ3 integrin. In vivo, absence or inhibition of PI3Kα resulted in a modest but significant decrease in thrombus size after superficial injuries of mouse mesenteric arteries and an increased time to arterial occlusion after carotid lesion, without modification in the tail bleeding time. Considering the more discrete and nonredundant role of PI3Kα compared with PI3Kβ, selective PI3Kα inhibitors are unlikely to increase the bleeding risk at least in the absence of combination with antiplatelet drugs or thrombopenia. Conclusions- This study provides mechanistic insight into the role of PI3Kα in platelet activation and arterial thrombosis.
Collapse
Affiliation(s)
- Pierre-Alexandre Laurent
- From the INSERM, UMR-S1048, Université Toulouse III, France (P.-A.L., R.S., A.R., C.C., T.A., S.S., B.P., M.-P.G.)
| | - Béatrice Hechler
- INSERM, EFS Grand Est, BPPS UMR-S 949, FMTS, Université de Strasbourg, France (B.H., P.H.M.)
| | - Romain Solinhac
- From the INSERM, UMR-S1048, Université Toulouse III, France (P.-A.L., R.S., A.R., C.C., T.A., S.S., B.P., M.-P.G.)
| | - Ashraf Ragab
- From the INSERM, UMR-S1048, Université Toulouse III, France (P.-A.L., R.S., A.R., C.C., T.A., S.S., B.P., M.-P.G.)
| | - Cendrine Cabou
- From the INSERM, UMR-S1048, Université Toulouse III, France (P.-A.L., R.S., A.R., C.C., T.A., S.S., B.P., M.-P.G.)
| | - Typhaine Anquetil
- From the INSERM, UMR-S1048, Université Toulouse III, France (P.-A.L., R.S., A.R., C.C., T.A., S.S., B.P., M.-P.G.)
| | - Sonia Severin
- From the INSERM, UMR-S1048, Université Toulouse III, France (P.-A.L., R.S., A.R., C.C., T.A., S.S., B.P., M.-P.G.)
| | - Cécile V Denis
- INSERM, UMR-S 1176, University of Paris-Sud, Université Paris-Saclay, France (C.V.D.)
| | - Pierre H Mangin
- INSERM, EFS Grand Est, BPPS UMR-S 949, FMTS, Université de Strasbourg, France (B.H., P.H.M.)
| | - Bart Vanhaesebroeck
- Cell Signaling, UCL Cancer Institute, University College London, United Kingdom (B.V.)
| | - Bernard Payrastre
- From the INSERM, UMR-S1048, Université Toulouse III, France (P.-A.L., R.S., A.R., C.C., T.A., S.S., B.P., M.-P.G.)
- CHU de Toulouse, Laboratoire d'Hématologie, France (B.P.)
| | - Marie-Pierre Gratacap
- From the INSERM, UMR-S1048, Université Toulouse III, France (P.-A.L., R.S., A.R., C.C., T.A., S.S., B.P., M.-P.G.)
| |
Collapse
|
42
|
Blair TA, Moore SF, Walsh TG, Hutchinson JL, Durrant TN, Anderson KE, Poole AW, Hers I. Phosphoinositide 3-kinase p110α negatively regulates thrombopoietin-mediated platelet activation and thrombus formation. Cell Signal 2018; 50:111-120. [PMID: 29793021 DOI: 10.1016/j.cellsig.2018.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 01/21/2023]
Abstract
Phosphoinositide 3-kinase (PI3K) plays an important role in platelet function and contributes to platelet hyperreactivity induced by elevated levels of circulating peptide hormones, including thrombopoietin (TPO). Previous work established an important role for the PI3K isoform; p110β in platelet function, however the role of p110α is still largely unexplored. Here we sought to investigate the role of p110α in TPO-mediated hyperactivity by using a conditional p110α knockout (KO) murine model in conjunction with platelet functional assays. We found that TPO-mediated enhancement of collagen-related peptide (CRP-XL)-induced platelet aggregation and adenosine triphosphate (ATP) secretion were significantly increased in p110α KO platelets. Furthermore, TPO-mediated enhancement of thrombus formation by p110α KO platelets was elevated over wild-type (WT) platelets, suggesting that p110α negatively regulates TPO-mediated priming of platelet function. The enhancements were not due to increased flow through the PI3K pathway as phosphatidylinositol 3,4,5-trisphosphate (PI(3,4,5)P3) formation and phosphorylation of Akt and glycogen synthase kinase 3 (GSK3) were comparable between WT and p110α KO platelets. In contrast, extracellular responsive kinase (ERK) phosphorylation and thromboxane (TxA2) formation were significantly enhanced in p110α KO platelets, both of which were blocked by the MEK inhibitor PD184352, whereas the p38 MAPK inhibitor VX-702 and p110α inhibitor PIK-75 had no effect. Acetylsalicylic acid (ASA) blocked the enhancement of thrombus formation by TPO in both WT and p110α KO mice. Together, these results demonstrate that p110α negatively regulates TPO-mediated enhancement of platelet function by restricting ERK phosphorylation and TxA2 synthesis in a manner independent of its kinase activity.
Collapse
Affiliation(s)
- T A Blair
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - S F Moore
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - T G Walsh
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - J L Hutchinson
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - T N Durrant
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - K E Anderson
- Inositide Laboratory, Babraham Institute, Cambridge, United Kingdom
| | - A W Poole
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - I Hers
- School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom.
| |
Collapse
|
43
|
The importance of blood platelet lipid signaling in thrombosis and in sepsis. Adv Biol Regul 2017; 67:66-73. [PMID: 28993230 DOI: 10.1016/j.jbior.2017.09.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 09/25/2017] [Accepted: 09/26/2017] [Indexed: 12/11/2022]
Abstract
Blood platelets are the first line of defense against hemorrhages and are also strongly involved in the processes of arterial thrombosis, a leading cause of death worldwide. Besides their well-established roles in hemostasis, vascular wall repair and thrombosis, platelets are now recognized as important players in other processes such as inflammation, healing, lymphangiogenesis, neoangiogenesis or cancer. Evidence is accumulating they are key effector cells in immune and inflammatory responses to host infection. To perform their different functions platelets express a wide variety of membrane receptors triggering specific intracellular signaling pathways and largely use lipid signaling systems. Lipid metabolism is highly active in stimulated platelets including the phosphoinositide metabolism with the phospholipase C (PLC) and the phosphoinositide 3-kinase (PI3K) pathways but also other enzymatic systems producing phosphatidic acid, lysophosphatidic acid, platelet activating factor, sphingosine 1-phosphate and a number of eicosanoids. While several of these bioactive lipids regulate intracellular platelet signaling mechanisms others are released by activated platelets acting as autocrine and/or paracrine factors modulating neighboring cells such as endothelial and immune cells. These bioactive lipids have been shown to play important roles in hemostasis and thrombosis but also in vessel integrity and dynamics, inflammation, tissue remodeling and wound healing. In this review, we will discuss some important aspects of platelet lipid signaling in thrombosis and during sepsis that is an important cause of death in intensive care unit. We will particularly focus on the implication of the different isoforms of PI3Ks and on the generation of eicosanoids released by activated platelets.
Collapse
|
44
|
Battram AM, Durrant TN, Agbani EO, Heesom KJ, Paul DS, Piatt R, Poole AW, Cullen PJ, Bergmeier W, Moore SF, Hers I. The Phosphatidylinositol 3,4,5-trisphosphate (PI(3,4,5)P3) Binder Rasa3 Regulates Phosphoinositide 3-kinase (PI3K)-dependent Integrin αIIbβ3 Outside-in Signaling. J Biol Chem 2017; 292:1691-1704. [PMID: 27903653 PMCID: PMC5290945 DOI: 10.1074/jbc.m116.746867] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 11/14/2016] [Indexed: 11/16/2022] Open
Abstract
The class I PI3K family of lipid kinases plays an important role in integrin αIIbβ3 function, thereby supporting thrombus growth and consolidation. Here, we identify Ras/Rap1GAP Rasa3 (GAP1IP4BP) as a major phosphatidylinositol 3,4,5-trisphosphate-binding protein in human platelets and a key regulator of integrin αIIbβ3 outside-in signaling. We demonstrate that cytosolic Rasa3 translocates to the plasma membrane in a PI3K-dependent manner upon activation of human platelets. Expression of wild-type Rasa3 in integrin αIIbβ3-expressing CHO cells blocked Rap1 activity and integrin αIIbβ3-mediated spreading on fibrinogen. In contrast, Rap1GAP-deficient (P489V) and Ras/Rap1GAP-deficient (R371Q) Rasa3 had no effect. We furthermore show that two Rasa3 mutants (H794L and G125V), which are expressed in different mouse models of thrombocytopenia, lack both Ras and Rap1GAP activity and do not affect integrin αIIbβ3-mediated spreading of CHO cells on fibrinogen. Platelets from thrombocytopenic mice expressing GAP-deficient Rasa3 (H794L) show increased spreading on fibrinogen, which in contrast to wild-type platelets is insensitive to PI3K inhibitors. Together, these results support an important role for Rasa3 in PI3K-dependent integrin αIIbβ3-mediated outside-in signaling and cell spreading.
Collapse
Affiliation(s)
- Anthony M Battram
- From the School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Tom N Durrant
- From the School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Ejaife O Agbani
- From the School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Kate J Heesom
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - David S Paul
- the McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514
| | - Raymond Piatt
- the McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514
| | - Alastair W Poole
- From the School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Peter J Cullen
- School of Biochemistry, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Wolfgang Bergmeier
- the McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514; Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514
| | - Samantha F Moore
- From the School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Ingeborg Hers
- From the School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, United Kingdom.
| |
Collapse
|
45
|
Downstream Regulatory Element Antagonist Modulator (DREAM), a target for anti-thrombotic agents. Pharmacol Res 2017; 117:283-287. [PMID: 28065857 DOI: 10.1016/j.phrs.2017.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/03/2017] [Indexed: 11/21/2022]
Abstract
Circulating platelets participate in the process of numerous diseases including thrombosis, inflammation, and cancer. Thus, it is of great importance to understand the underlying mechanisms mediating platelet activation under disease conditions. Emerging evidence indicates that despite the lack of a nucleus, platelets possess molecules that are involved in gene transcription in nucleated cells. This review will summarize downstream regulatory element antagonist modulator (DREAM), a transcriptional repressor, and highlight recent findings suggesting its novel non-transcriptional role in hemostasis and thrombosis.
Collapse
|
46
|
DREAM plays an important role in platelet activation and thrombogenesis. Blood 2016; 129:209-225. [PMID: 27903531 DOI: 10.1182/blood-2016-07-724419] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 11/23/2016] [Indexed: 01/18/2023] Open
Abstract
Downstream regulatory element antagonist modulator (DREAM), a transcriptional repressor, is known to modulate pain responses. However, it is unknown whether DREAM is expressed in anucleate platelets and plays a role in thrombogenesis. By using intravital microscopy with DREAM-null mice and their bone marrow chimeras, we demonstrated that both hematopoietic and nonhematopoietic cell DREAMs are required for platelet thrombus formation following laser-induced arteriolar injury. In a FeCl3-induced thrombosis model, we found that compared with wild-type (WT) control and nonhematopoietic DREAM knockout (KO) mice, DREAM KO control and hematopoietic DREAM KO mice showed a significant delay in time to occlusion. Tail bleeding time was prolonged in DREAM KO control mice, but not in WT or DREAM bone marrow chimeric mice. In vivo adoptive transfer experiments further indicated the importance of platelet DREAM in thrombogenesis. We found that DREAM deletion does not alter the ultrastructural features of platelets but significantly impairs platelet aggregation and adenosine triphosphate secretion induced by numerous agonists (collagen-related peptide, adenosine 5'-diphosphate, A23187, thrombin, or U46619). Biochemical studies revealed that platelet DREAM positively regulates phosphoinositide 3-kinase (PI3K) activity during platelet activation. Using DREAM-null platelets and PI3K isoform-specific inhibitors, we observed that platelet DREAM is important for α-granule secretion, Ca2+ mobilization, and aggregation through PI3K class Iβ (PI3K-Iβ). Genetic and pharmacological studies in human megakaryoblastic MEG-01 cells showed that DREAM is important for A23187-induced Ca2+ mobilization and its regulatory function requires Ca2+ binding and PI3K-Iβ activation. These results suggest that platelet DREAM regulates PI3K-Iβ activity and plays an important role during thrombus formation.
Collapse
|
47
|
Van Aelst B, Devloo R, Zachée P, t'Kindt R, Sandra K, Vandekerckhove P, Compernolle V, Feys HB. Psoralen and Ultraviolet A Light Treatment Directly Affects Phosphatidylinositol 3-Kinase Signal Transduction by Altering Plasma Membrane Packing. J Biol Chem 2016; 291:24364-24376. [PMID: 27687726 DOI: 10.1074/jbc.m116.735126] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 09/17/2016] [Indexed: 01/15/2023] Open
Abstract
Psoralen and ultraviolet A light (PUVA) are used to kill pathogens in blood products and as a treatment of aberrant cell proliferation in dermatitis, cutaneous T-cell lymphoma, and graft-versus-host disease. DNA damage is well described, but the direct effects of PUVA on cell signal transduction are poorly understood. Because platelets are anucleate and contain archetypal signal transduction machinery, they are ideally suited to address this. Lipidomics on platelet membrane extracts showed that psoralen forms adducts with unsaturated carbon bonds of fatty acyls in all major phospholipid classes after PUVA. Such adducts increased lipid packing as measured by a blue shift of an environment-sensitive fluorescent probe in model liposomes. Furthermore, the interaction of these liposomes with lipid order-sensitive proteins like amphipathic lipid-packing sensor and α-synuclein was inhibited by PUVA. In platelets, PUVA caused poor membrane binding of Akt and Bruton's tyrosine kinase effectors following activation of the collagen glycoprotein VI and thrombin protease-activated receptor (PAR) 1. This resulted in defective Akt phosphorylation despite unaltered phosphatidylinositol 3,4,5-trisphosphate levels. Downstream integrin activation was furthermore affected similarly by PUVA following PAR1 (effective half-maximal concentration (EC50), 8.4 ± 1.1 versus 4.3 ± 1.1 μm) and glycoprotein VI (EC50, 1.61 ± 0.85 versus 0.26 ± 0.21 μg/ml) but not PAR4 (EC50, 50 ± 1 versus 58 ± 1 μm) signal transduction. Our findings were confirmed in T-cells from graft-versus-host disease patients treated with extracorporeal photopheresis, a form of systemic PUVA. In conclusion, PUVA increases the order of lipid phases by covalent modification of phospholipids, thereby inhibiting membrane recruitment of effector kinases.
Collapse
Affiliation(s)
- Britt Van Aelst
- From the Transfusion Research Center, Belgian Red Cross-Flanders, 9000 Ghent, Belgium
| | - Rosalie Devloo
- From the Transfusion Research Center, Belgian Red Cross-Flanders, 9000 Ghent, Belgium
| | - Pierre Zachée
- the Department of Hematology, Hospital Network Antwerp, 2000 Antwerp, Belgium
| | - Ruben t'Kindt
- the Research Institute for Chromatography, 8500 Kortrijk, Belgium
| | - Koen Sandra
- the Research Institute for Chromatography, 8500 Kortrijk, Belgium
| | - Philippe Vandekerckhove
- the Blood Service of the Belgian Red Cross-Flanders, 2800 Mechelen, Belgium,; the Department of Public Health and Primary Care, KULeuven, 3000 Leuven, Belgium, and; the Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Veerle Compernolle
- From the Transfusion Research Center, Belgian Red Cross-Flanders, 9000 Ghent, Belgium,; the Blood Service of the Belgian Red Cross-Flanders, 2800 Mechelen, Belgium,; the Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Hendrik B Feys
- From the Transfusion Research Center, Belgian Red Cross-Flanders, 9000 Ghent, Belgium,.
| |
Collapse
|
48
|
Münzer P, Walker-Allgaier B, Geue S, Geuss E, Hron G, Rath D, Eißler D, Winter S, Schaeffeler E, Meinert M, Schaller M, Greinacher A, Schwab M, Geisler T, Kleinschnitz C, Lang F, Gawaz M, Borst O. PDK1 Determines Collagen-Dependent Platelet Ca
2+
Signaling and Is Critical to Development of Ischemic Stroke In Vivo. Arterioscler Thromb Vasc Biol 2016; 36:1507-16. [DOI: 10.1161/atvbaha.115.307105] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 05/16/2016] [Indexed: 01/12/2023]
Abstract
Objective—
Activation of platelets by subendothelial collagen results in an increase of cytosolic Ca
2+
concentration ([Ca
2+
]
i
) and is followed by platelet activation and thrombus formation that may lead to vascular occlusion. The present study determined the role of phosphoinositide-dependent protein kinase 1 (PDK1) in collagen-dependent platelet Ca
2+
signaling and ischemic stroke in vivo.
Approach and Results—
Platelet activation with collagen receptor glycoprotein VI agonists collagen-related peptide or convulxin resulted in a significant increase in PDK1 activity independent of second-wave signaling. PDK1 deficiency was associated with reduced platelet phospholipase Cγ2–dependent inositol-1,4,5-trisphosphate production and intracellular [Ca
2+
]
i
in response to stimulation with collagen-related peptide or convulxin. The defective increase of [Ca
2+
]
i
resulted in a substantial defect in activation-dependent platelet secretion and aggregation on collagen-related peptide stimulation. Furthermore, Rac1 activation and spreading, adhesion to collagen, and thrombus formation under high arterial shear rates were significantly diminished in PDK1-deficient platelets. Mice with PDK1-deficient platelets were protected against arterial thrombotic occlusion after FeCl
3
-induced mesenteric arterioles injury and ischemic stroke in vivo. These mice had significantly reduced brain infarct volumes, with a significantly increased survival of 7 days after transient middle cerebral artery occlusion without increase of intracerebral hemorrhage. Tail bleeding time was prolonged in
pdk1
−/−
mice, reflecting an important role of PDK1 in primary hemostasis.
Conclusions—
PDK1 is required for Ca
2+
-dependent platelet activation on stimulation of collagen receptor glycoprotein VI, arterial thrombotic occlusion, and ischemic stroke in vivo.
Collapse
Affiliation(s)
- Patrick Münzer
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Britta Walker-Allgaier
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Sascha Geue
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Eva Geuss
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Gregor Hron
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Dominik Rath
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Daniela Eißler
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Stefan Winter
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Elke Schaeffeler
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Monika Meinert
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Martin Schaller
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Andreas Greinacher
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Matthias Schwab
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Tobias Geisler
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Christoph Kleinschnitz
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Florian Lang
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Meinrad Gawaz
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| | - Oliver Borst
- From the Department of Cardiology and Cardiovascular Medicine (P.M., B.W.-A., S.G., D.R., D.E., T.G., M.G., O.B.), Department of Physiology (B.W.-A., F.L.), Department of Evolutionary Biology of Invertebrates, Institute for Evolution and Ecology (M.M.), Department of Dermatology (M.S.), and Department of Clinical Pharmacology (M.S.), University of Tübingen, Tübingen, Germany; Department of Neurology, University of Würzburg, Würzburg, Germany (E.G., C.K.); Institute for Immunology and Transfusion
| |
Collapse
|
49
|
Bye AP, Unsworth AJ, Gibbins JM. Platelet signaling: a complex interplay between inhibitory and activatory networks. J Thromb Haemost 2016; 14:918-30. [PMID: 26929147 PMCID: PMC4879507 DOI: 10.1111/jth.13302] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 02/11/2016] [Indexed: 01/22/2023]
Abstract
The role of platelets in hemostasis and thrombosis is dependent on a complex balance of activatory and inhibitory signaling pathways. Inhibitory signals released from the healthy vasculature suppress platelet activation in the absence of platelet receptor agonists. Activatory signals present at a site of injury initiate platelet activation and thrombus formation; subsequently, endogenous negative signaling regulators dampen activatory signals to control thrombus growth. Understanding the complex interplay between activatory and inhibitory signaling networks is an emerging challenge in the study of platelet biology, and necessitates a systematic approach to utilize experimental data effectively. In this review, we will explore the key points of platelet regulation and signaling that maintain platelets in a resting state, mediate activation to elicit thrombus formation, or provide negative feedback. Platelet signaling will be described in terms of key signaling molecules that are common to the pathways activated by platelet agonists and can be described as regulatory nodes for both positive and negative regulators.
Collapse
Affiliation(s)
- A P Bye
- Institute of Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK
| | - A J Unsworth
- Institute of Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK
| | - J M Gibbins
- Institute of Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK
| |
Collapse
|
50
|
Mattheij NJA, Braun A, van Kruchten R, Castoldi E, Pircher J, Baaten CCFMJ, Wülling M, Kuijpers MJE, Köhler R, Poole AW, Schreiber R, Vortkamp A, Collins PW, Nieswandt B, Kunzelmann K, Cosemans JMEM, Heemskerk JWM. Survival protein anoctamin-6 controls multiple platelet responses including phospholipid scrambling, swelling, and protein cleavage. FASEB J 2016; 30:727-37. [PMID: 26481309 DOI: 10.1096/fj.15-280446] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 10/05/2015] [Indexed: 11/11/2022]
Abstract
Scott syndrome is a rare bleeding disorder, characterized by altered Ca(2+)-dependent platelet signaling with defective phosphatidylserine (PS) exposure and microparticle formation, and is linked to mutations in the ANO6 gene, encoding anoctamin (Ano)6. We investigated how the complex platelet phenotype of this syndrome is linked to defective expression of Anos or other ion channels. Mice were generated with heterozygous of homozygous deficiency in Ano6, Ano1, or Ca(2+)-dependent KCa3.1 Gardos channel. Platelets from these mice were extensively analyzed on molecular functions and compared with platelets from a patient with Scott syndrome. Deficiency in Ano1 or Gardos channel did not reduce platelet responses compared with control mice (P > 0.1). In 2 mouse strains, deficiency in Ano6 resulted in reduced viability with increased bleeding time to 28.6 min (control 6.4 min, P < 0.05). Platelets from the surviving Ano6-deficient mice resembled platelets from patients with Scott syndrome in: 1) normal collagen-induced aggregate formation (P > 0.05) with reduced PS exposure (-65 to 90%); 2) lowered Ca(2+)-dependent swelling (-80%) and membrane blebbing (-90%); 3) reduced calpain-dependent protein cleavage (-60%); and 4) moderately affected apoptosis-dependent PS exposure. In conclusion, mouse deficiency of Ano6 but not of other channels affects viability and phenocopies the complex changes in platelets from hemostatically impaired patients with Scott syndrome.
Collapse
Affiliation(s)
- Nadine J A Mattheij
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Attila Braun
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Roger van Kruchten
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Elisabetta Castoldi
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Joachim Pircher
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Constance C F M J Baaten
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Manuela Wülling
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Marijke J E Kuijpers
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Ralf Köhler
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Alastair W Poole
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Rainer Schreiber
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Andrea Vortkamp
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Peter W Collins
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Bernhard Nieswandt
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Karl Kunzelmann
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Judith M E M Cosemans
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Johan W M Heemskerk
- *Department of Cell Biochemistry of Thrombosis and Haemostasis Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, The Netherlands; Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; Walter Brendel Centre of Experimental Medicine and German Centre of Cardiovascular Research, Munich Heart Alliance, Ludwig-Maximilians-Universität München, München, Germany; Department of Developmental Biology, Centre for Medical Biotechnology, University of Duisburg-Essen, Duisburg-Essen, Germany; Aragon Institute of Health Sciences I+CS/IIS and ARAID, Zaragoza, Spain; School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom; Institute of Physiology, University of Regensburg, Regensburg, Germany; **Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|