101
|
Ngo T, Kim K, Bian Y, Noh H, Lim KM, Chung JH, Bae ON. Antithrombotic Effects of Paeoniflorin from Paeonia suffruticosa by Selective Inhibition on Shear Stress-Induced Platelet Aggregation. Int J Mol Sci 2019; 20:ijms20205040. [PMID: 31614534 PMCID: PMC6834133 DOI: 10.3390/ijms20205040] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/30/2019] [Accepted: 10/09/2019] [Indexed: 12/16/2022] Open
Abstract
Antiplatelet agents are important in the pharmacotherapeutic regime for many cardiovascular diseases, including thrombotic disorders. However, bleeding, the most serious adverse effect associated with current antiplatelet therapy, has led to many efforts to discover novel anti-platelet drugs without bleeding issues. Of note, shear stress-induced platelet aggregation (SIPA) is a promising target to overcome bleeding since SIPA happens only in pathological conditions. Accordingly, this study was carried out to discover antiplatelet agents selectively targeting SIPA. By screening various herbal extracts, Paeonia suffruticosa and its major bioactive constituent, paeoniflorin, were identified to have significant inhibitory effects against shear-induced aggregation in human platelets. The effects of paeoniflorin on intraplatelet calcium levels, platelet degranulation, and integrin activation in high shear stress conditions were evaluated by a range of in vitro experiments using human platelets. The inhibitory effect of paeoniflorin was determined to be highly selective against SIPA, through modulating von Willebrand Factor (vWF)-platelet glycoprotein Ib (GP Ib) interaction. The effects of paeoniflorin on platelet functions under high shear stress were confirmed in the ex vivo SIPA models in rats, showing the good accordance with the anti-SIPA effects on human platelets. Treatment with paeoniflorin significantly prevented arterial thrombosis in vivo from the dose of 10 mg/kg without prolonging bleeding time or blood clotting time in rats. Collectively, our results demonstrated that paeoniflorin can be a novel anti-platelet agent selectively targeting SIPA with an improved safety profile.
Collapse
Affiliation(s)
- Thien Ngo
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
- Faculty of Pharmacy, Thai Binh University of Medicine and Pharmacy, Thai Binh city 410000, Vietnam.
| | - Keunyoung Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Yiying Bian
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
- School of Public Health, China Medical University, Shenyang 110122, China.
| | - Hakjun Noh
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Kyung-Min Lim
- College of Pharmacy, Ewha Womans University, Seoul 03760, Korea.
| | - Jin-Ho Chung
- College of Pharmacy, Seoul National University, Seoul 08826, Korea.
| | - Ok-Nam Bae
- College of Pharmacy, Hanyang University, Ansan 15588, Korea.
| |
Collapse
|
102
|
Bellio M, Caux M, Vauclard A, Chicanne G, Gratacap MP, Terrisse AD, Severin S, Payrastre B. Phosphatidylinositol 3 monophosphate metabolizing enzymes in blood platelet production and in thrombosis. Adv Biol Regul 2019; 75:100664. [PMID: 31604685 DOI: 10.1016/j.jbior.2019.100664] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/19/2019] [Accepted: 09/30/2019] [Indexed: 02/09/2023]
Abstract
Blood platelets, produced by the fragmentation of megakaryocytes, play a key role in hemostasis and thrombosis. Being implicated in atherothrombosis and other thromboembolic disorders, they represent a major therapeutic target for antithrombotic drug development. Several recent studies have highlighted an important role for the lipid phosphatidylinositol 3 monophosphate (PtdIns3P) in megakaryocytes and platelets. PtdIns3P, present in small amounts in mammalian cells, is involved in the control of endocytic trafficking and autophagy. Its metabolism is finely regulated by specific kinases and phosphatases. Class II (α, β and γ) and III (Vps34) phosphoinositide-3-kinases (PI3Ks), INPP4 and Fig4 are involved in the production of PtdIns3P whereas PIKFyve, myotubularins (MTMs) and type II PIPK metabolize PtdIns3P. By regulating the turnover of different pools of PtdIns3P, class II (PI3KC2α) and class III (Vps34) PI3Ks have been recently involved in the regulation of platelet production and functions. These pools of PtdIns3P appear to modulate membrane organization and intracellular trafficking. Moreover, PIKFyve and INPP4 have been recently implicated in arterial thrombosis. In this review, we will discuss the role of PtdIns3P metabolizing enzymes in platelet production and function. Potential new anti-thrombotic therapeutic perspectives based on inhibitors targeting specifically PtdIns3P metabolizing enzymes will also be commented.
Collapse
Affiliation(s)
- Marie Bellio
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Manuella Caux
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Alicia Vauclard
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Gaëtan Chicanne
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Marie-Pierre Gratacap
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Anne-Dominique Terrisse
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Sonia Severin
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - Bernard Payrastre
- Inserm U1048 and Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France; Laboratoire d'Hématologie, Hopital Universitaire de Toulouse, Toulouse, France.
| |
Collapse
|
103
|
Ngo T, Kim K, Bian Y, An GJ, Bae ON, Lim KM, Chung JH. Cyclocurcumin from Curcuma longa selectively inhibits shear stress-induced platelet aggregation. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103462] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
104
|
Kannan M, Ahmad F, Saxena R. Platelet activation markers in evaluation of thrombotic risk factors in various clinical settings. Blood Rev 2019; 37:100583. [DOI: 10.1016/j.blre.2019.05.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 05/11/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022]
|
105
|
Abstract
Dysregulation of lymphocyte function, accumulation of autoantibodies and defective clearance of circulating immune complexes and apoptotic cells are hallmarks of systemic lupus erythematosus (SLE). Moreover, it is now evident that an intricate interplay between the adaptive and innate immune systems contributes to the pathogenesis of SLE, ultimately resulting in chronic inflammation and organ damage. Platelets circulate in the blood and are chiefly recognized for their role in the prevention of bleeding and promotion of haemostasis; however, accumulating evidence points to a role for platelets in both adaptive and innate immunity. Through a broad repertoire of receptors, platelets respond promptly to immune complexes, complement and damage-associated molecular patterns, and represent a major reservoir of immunomodulatory molecules in the circulation. Furthermore, evidence suggests that platelets are activated in patients with SLE, and that they could contribute to the circulatory autoantigenic load through the release of microparticles and mitochondrial antigens. Herein, we highlight how platelets contribute to the immune response and review evidence implicating platelets in the pathogenesis of SLE.
Collapse
|
106
|
Kerrigan SW, Devine T, Fitzpatrick G, Thachil J, Cox D. Early Host Interactions That Drive the Dysregulated Response in Sepsis. Front Immunol 2019; 10:1748. [PMID: 31447831 PMCID: PMC6691039 DOI: 10.3389/fimmu.2019.01748] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/10/2019] [Indexed: 01/18/2023] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. While many individual cells and systems in the body are involved in driving the excessive and sometimes sustained host response, pathogen engagement with endothelial cells and platelets early in sepsis progression, are believed to be key. Significant progress has been made in establishing key molecular interactions between platelets and pathogens and endothelial cells and pathogens. This review will explore the growing number of compensatory connections between bacteria and viruses with platelets and endothelial cells and how a better understanding of these interactions are informing the field of potential novel ways to treat the dysregulated host response during sepsis.
Collapse
Affiliation(s)
- Steven W Kerrigan
- Cardiovascular Infection Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland.,Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tatyana Devine
- Cardiovascular Infection Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Glenn Fitzpatrick
- Cardiovascular Infection Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jecko Thachil
- Department of Haematology, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Dermot Cox
- Cardiovascular Infection Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
107
|
Stainer AR, Sasikumar P, Bye AP, Unsworth AJ, Holbrook LM, Tindall M, Lovegrove JA, Gibbins JM. The Metabolites of the Dietary Flavonoid Quercetin Possess Potent Antithrombotic Activity, and Interact with Aspirin to Enhance Antiplatelet Effects. TH OPEN 2019; 3:e244-e258. [PMID: 31367693 PMCID: PMC6667742 DOI: 10.1055/s-0039-1694028] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 06/24/2019] [Indexed: 01/09/2023] Open
Abstract
Quercetin, a dietary flavonoid, has been reported to possess antiplatelet activity. However, its extensive metabolism following ingestion has resulted in difficulty elucidating precise mechanisms of action. In this study, we aimed to characterize the antiplatelet mechanisms of two methylated metabolites of quercetin-isorhamnetin and tamarixetin-and explore potential interactions with aspirin. Isorhamnetin and tamarixetin inhibited human platelet aggregation, and suppressed activatory processes including granule secretion, integrin αIIbβ3 function, calcium mobilization, and spleen tyrosine kinase (Syk)/linker for activation of T cells (LAT) phosphorylation downstream of glycoprotein VI with similar potency to quercetin. All three flavonoids attenuated thrombus formation in an in vitro microfluidic model, and isoquercetin, a 3-O-glucoside of quercetin, inhibited thrombosis in a murine laser injury model. Isorhamnetin, tamarixetin, and quercetin enhanced the antiplatelet effects of aspirin more-than-additively in a plate-based aggregometry assay, reducing aspirin IC 50 values by an order of magnitude, with this synergy maintained in a whole blood test of platelet function. Our data provide mechanistic evidence for the antiplatelet activity of two quercetin metabolites, isorhamnetin and tamarixetin, and suggest a potential antithrombotic role for these flavonoids. In combination with their interactions with aspirin, this may represent a novel avenue of investigation for the development of new antithrombotic strategies and management of current therapies.
Collapse
Affiliation(s)
- Alexander R Stainer
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Parvathy Sasikumar
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom.,Centre for Haematology, Imperial College London, London, United Kingdom
| | - Alexander P Bye
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Amanda J Unsworth
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom.,School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
| | - Lisa M Holbrook
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom.,School of Cardiovascular Medicine and Sciences, King's College London, London, United Kingdom
| | - Marcus Tindall
- Department of Mathematics and Statistics, University of Reading, Reading, United Kingdom
| | - Julie A Lovegrove
- Department of Food and Nutritional Sciences, Hugh Sinclair Unit of Human Nutrition, University of Reading, Reading, United Kingdom
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, United Kingdom
| |
Collapse
|
108
|
Nurden AT. Clinical significance of altered collagen-receptor functioning in platelets with emphasis on glycoprotein VI. Blood Rev 2019; 38:100592. [PMID: 31351674 DOI: 10.1016/j.blre.2019.100592] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/05/2019] [Accepted: 07/19/2019] [Indexed: 01/01/2023]
Abstract
Much interest surrounds the receptors α2β1 and glycoprotein VI (GPVI) whose synchronized action mediates the attachment and activation of platelets on collagen, essential for preventing blood loss but also the most thrombogenic component of the vessel wall. Subject to density variations on platelets through natural polymorphisms, the absence of α2β1 or GPVI uniquely leads to a substantial block of hemostasis without causing major bleeding. Specific to the megakaryocyte lineage, GPVI and its signaling pathways are most promising targets for anti-thrombotic therapy. This review looks at the clinical consequences of the loss of collagen receptor function with emphasis on both the inherited and acquired loss of GPVI with brief mention of mouse models when necessary. A detailed survey of rare case reports of patients with inherited disease-causing variants of the GP6 gene is followed by an assessment of the causes and clinical consequences of acquired GPVI deficiency, a more frequent finding most often due to antibody-induced platelet GPVI shedding. Release of soluble GPVI is brought about by platelet metalloproteinases; a process induced by ligand or antibody binding to GPVI or even high shear forces. Also included is an assessment of the clinical importance of GPVI-mediated platelet interactions with fibrin and of the promise shown by the pharmacological inhibition of GPVI in a cardiovascular context. The role for GPVI in platelet function in inflammation and in the evolution and treatment of major illnesses such as rheumatoid arthritis, cancer and sepsis is also discussed.
Collapse
Affiliation(s)
- Alan T Nurden
- Institut de Rhythmologie et de Modélisation Cardiaque, PTIB, Hôpital Xavier Arnozan, 33600 Pessac, France.
| |
Collapse
|
109
|
Walsh TG, Wersäll A, Poole AW. Characterisation of the Ral GTPase inhibitor RBC8 in human and mouse platelets. Cell Signal 2019; 59:34-40. [PMID: 30880223 PMCID: PMC6510928 DOI: 10.1016/j.cellsig.2019.03.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/13/2019] [Accepted: 03/13/2019] [Indexed: 01/28/2023]
Abstract
The Ral GTPases, RalA and RalB, have been implicated in numerous cellular processes, but are most widely known for having regulatory roles in exocytosis. Recently, we demonstrated that deletion of both Ral genes in a platelet-specific mouse gene knockout caused a substantial defect in surface exposure of P-selectin, with only a relatively weak defect in platelet dense granule secretion that did not alter platelet functional responses such as aggregation or thrombus formation. We sought to investigate the function of Rals in human platelets using the recently described Ral inhibitor, RBC8. Initial studies in human platelets confirmed that RBC8 could effectively inhibit Ral GTPase activation, with an IC50 of 2.2 μM and 2.3 μM for RalA and RalB, respectively. Functional studies using RBC8 revealed significant, dose-dependent inhibition of platelet aggregation, secretion (α- and dense granule), integrin activation and thrombus formation, while α-granule release of platelet factor 4, Ca2+ signalling or phosphatidylserine exposure were unaltered. Subsequent studies in RalAB-null mouse platelets pretreated with RBC8 showed dose-dependent decreases in integrin activation and dense granule secretion, with significant inhibition of platelet aggregation and P-selectin exposure at 10 μM RBC8. This study strongly suggests therefore that although RBC8 is useful as a Ral inhibitor in platelets, it is likely also to have off-target effects in the same concentration range as for Ral inhibition. So, whilst clearly useful as a Ral inhibitor, interpretation of data needs to take this into account when assessing roles for Rals using RBC8.
Collapse
Affiliation(s)
- Tony G Walsh
- From the School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol BS8 1TD, United Kingdom.
| | - Andreas Wersäll
- From the School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Alastair W Poole
- From the School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol BS8 1TD, United Kingdom
| |
Collapse
|
110
|
Villmann JM, Burkhardt R, Teren A, Villmann T, Thiery J, Drogies T. Atherosclerosis, myocardial infarction and primary hemostasis: Impact of platelets, von Willebrand factor and soluble glycoprotein VI. Thromb Res 2019; 180:98-104. [PMID: 31276978 DOI: 10.1016/j.thromres.2019.06.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/07/2019] [Accepted: 06/24/2019] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Little is known about peril constellations in primary hemostasis contributing to an acute myocardial infarction (MI) in patients with already manifest atherosclerosis. The study aimed to establish a predicting model based on six biomarkers of primary hemostasis: platelet count, mean platelet volume, hematocrit, soluble glycoprotein VI, fibrinogen and von Willebrand factor ratio. MATERIALS AND METHODS The biomarkers were measured in 1.491 patients with manifest atherosclerosis of the Leipzig (LIFE) heart study. Three groups were divided: patients with coronary artery disease (900 patients) and patients with atherosclerosis and either ST-elevated MI (404 patients) or Non-ST-elevated MI (187 patients). Correlations were analyzed by non-linear analysis with Self Organizing Maps. Classification and discriminant analysis was performed using Learning Vector Quantization. RESULTS AND CONCLUSIONS The combination of hemostatic biomarkers is regarded as valuable tool for identifying patients with atherosclerosis at risk for MI. Nevertheless, our study contradicts this belief. The biomarkers did not allow to establish a predicting model usable in daily patient care. Good specificity and sensitivity for the detection of MI was only reached in models including acute phase parameters (specificity 0,9036, sensitivity 0,7937 in men; 0,8977 and 0,8133 in women). In detail, hematocrit and soluble glycoprotein VI were significantly different between the groups. Significant dissimilarities were also found for fibrinogen (in men) and von Willebrand factor ratio. In contrast, the most promising parameters mean platelet volume and platelet count showed no difference, which is an important contribution to the controversy concerning them as new risk and therapy targets for MI.
Collapse
Affiliation(s)
- Josepha-Maria Villmann
- Institute for Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital of Leipzig, Leipzig, Germany; LIFE - Leipzig Research Center for Civilization Diseases, University Leipzig, Germany
| | - Ralph Burkhardt
- Institute for Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital of Leipzig, Leipzig, Germany
| | - Andrej Teren
- LIFE - Leipzig Research Center for Civilization Diseases, University Leipzig, Germany; Leipzig Heart Center, Leipzig, Germany
| | - Thomas Villmann
- Computational Intelligence Group, University of Applied Sciences Mittweida, Germany
| | - Joachim Thiery
- Institute for Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital of Leipzig, Leipzig, Germany; LIFE - Leipzig Research Center for Civilization Diseases, University Leipzig, Germany
| | - Tim Drogies
- Institute for Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital of Leipzig, Leipzig, Germany; LIFE - Leipzig Research Center for Civilization Diseases, University Leipzig, Germany.
| |
Collapse
|
111
|
Dey S, Lo HJ, Wong CH. An Efficient Modular One-Pot Synthesis of Heparin-Based Anticoagulant Idraparinux. J Am Chem Soc 2019; 141:10309-10314. [PMID: 31244187 DOI: 10.1021/jacs.9b03266] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Idraparinux is a fully O-sulfated α-methyl glycoside of heparin pentasaccharide motif known to interact with the antithrombin III domain and act as anticoagulant. The current most effective synthesis of Idraparinux is complicated and nonstereoselective, requiring numerous stepwise procedures with low yields. We report here an efficient modular one-pot synthesis of Idraparinux involving the use of a glycosyl phosphate with 6- O- tert-butyl diphenyl silyl group and a d-glucuronic acid-containing disaccharide thioglycoside with 6- O-acetyl group as donor building blocks for the α-directing one-pot glycosylations with an l-iduronic acid-containing disaccharide acceptor building block. The uronic acid was incorporated in a disaccharide module used in the one-pot synthesis to avoid the complicated late-stage installation of these acidic sugars. The one-pot synthesis of Idraparinux demonstrated here is an effective strategy and should be applicable to the modular assembly of other heparan sulfates with regiodefined sulfation pattern for functional study.
Collapse
Affiliation(s)
- Supriya Dey
- Department of Chemistry , Scripps Research , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Hong-Jay Lo
- Department of Chemistry , Scripps Research , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Chi-Huey Wong
- Department of Chemistry , Scripps Research , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States.,The Genomics Research Center , Academia Sinica , 128 Academia Road, Section 2 , Taipei 115 , Taiwan
| |
Collapse
|
112
|
Petrenko A, Carnevale M, Somov A, Osorio J, Rodríguez J, Guibert E, Fuller B, Froghi F. Organ Preservation into the 2020s: The Era of Dynamic Intervention. Transfus Med Hemother 2019; 46:151-172. [PMID: 31244584 PMCID: PMC6558325 DOI: 10.1159/000499610] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 02/04/2019] [Indexed: 12/12/2022] Open
Abstract
Organ preservation has been of major importance ever since transplantation developed into a global clinical activity. The relatively simple procedures were developed on a basic comprehension of low-temperature biology as related to organs outside the body. In the past decade, there has been a significant increase in knowledge of the sequelae of effects in preserved organs, and how dynamic intervention by perfusion can be used to mitigate injury and improve the quality of the donated organs. The present review focuses on (1) new information about the cell and molecular events impacting on ischemia/reperfusion injury during organ preservation, (2) strategies which use varied compositions and additives in organ preservation solutions to deal with these, (3) clear definitions of the developing protocols for dynamic organ perfusion preservation, (4) information on how the choice of perfusion solutions can impact on desired attributes of dynamic organ perfusion, and (5) summary and future horizons.
Collapse
Affiliation(s)
- Alexander Petrenko
- Department of Cryobiochemistry, Institute for Problems of Cryobiology and Cryomedicine, Ukraine Academy of Sciences, Kharkov, Ukraine
| | - Matias Carnevale
- Centro Binacional (Argentina-Italia) de Investigaciones en Criobiología Clínica y Aplicada (CAIC), Universidad Nacional de Rosario, Rosario, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Alexander Somov
- Department of Cryobiochemistry, Institute for Problems of Cryobiology and Cryomedicine, Ukraine Academy of Sciences, Kharkov, Ukraine
| | - Juliana Osorio
- Centro Binacional (Argentina-Italia) de Investigaciones en Criobiología Clínica y Aplicada (CAIC), Universidad Nacional de Rosario, Rosario, Argentina
| | - Joaquin Rodríguez
- Centro Binacional (Argentina-Italia) de Investigaciones en Criobiología Clínica y Aplicada (CAIC), Universidad Nacional de Rosario, Rosario, Argentina
| | - Edgardo Guibert
- Centro Binacional (Argentina-Italia) de Investigaciones en Criobiología Clínica y Aplicada (CAIC), Universidad Nacional de Rosario, Rosario, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Barry Fuller
- UCL Division of Surgery and Interventional Sciences, Royal Free Hospital, London, United Kingdom
| | - Farid Froghi
- UCL Division of Surgery and Interventional Sciences, Royal Free Hospital, London, United Kingdom
| |
Collapse
|
113
|
Staszewski J, Pogoda A, Data K, Walczak K, Nowocień M, Frankowska E, Stępień A. The mean platelet volume on admission predicts unfavorable stroke outcomes in patients treated with IV thrombolysis. Clin Interv Aging 2019; 14:493-503. [PMID: 30880930 PMCID: PMC6398411 DOI: 10.2147/cia.s195451] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose The role of biomarkers in the prediction of acute ischemic stroke (AIS) outcome or response to thrombolytic therapy (with recombinant tissue plasminogen activator [rt-PA]) remains limited. The aim of this study was to evaluate whether mean platelet volume (MPV) could predict short-term functional outcome in patients with AIS following rt-PA treatment. Patients and methods This was a retrospective analysis of 237 AIS patients (mean age 71.04±0.8 years, 50.6% women) consecutively admitted to a tertiary care center between 2011 and 2015. Results The mean MPV in the cohort was 9.8±0.35 fL (lowest tertile <7.29 fL, median 7.29–8.8 fL, and highest tertile >8.8 fL). Patients in the lowest tertile compared to median and highest tertiles were less often dependent (modified Rankin scale [mRS] ≥3) at admission (87.2% vs 96.1% and 96.1%, respectively, P=0.04) and less often had a poor stroke outcome (mRS 4–6) at discharge (28.2% vs 55.3% and 44.7%, P<0.01). However, there was no significant difference between tertiles with regard to AIS etiology, CT (Alberta Stroke Program Early CT) score, frequency of stroke due to large artery occlusion, risk of secondary hemorrhage, and early neurologic deterioration. Multivariable analysis after adjustment for confounders showed that patients in the second and third tertiles had a significantly higher risk of poor stroke outcome (OR =1.9, 95% CI =1.01–4), lack of early improvement (OR =1.91, 95% CI =1.05–3.47), lower chance of good outcome (mRS 0–2; OR =0.38, 95% CI =0.18–0.78), or minor stroke at discharge (OR =0.47, 95% CI =0.26–0.84). Receiver operating characteristic analysis for prediction of poor stroke outcome showed that the optimal cut-off point of MPV was 8.8 fL (area under the curve 0.586 [0.512–0.659], P=0.03) with a sensitivity of 82.7% and a specificity of 43.9%. Conclusion Disabling or fatal ischemic stroke in thrombolyzed patients was observed more often in patients with high admission MPV. The prognostic value of MPV was independent of other well-defined individual risk factors.
Collapse
Affiliation(s)
- Jacek Staszewski
- Clinic of Neurology, Military Institute of Medicine, Warsaw, Poland,
| | - Aleksandra Pogoda
- Clinic of Neurology, Military Institute of Medicine, Warsaw, Poland,
| | - Kamila Data
- Clinic of Neurology, Military Institute of Medicine, Warsaw, Poland,
| | - Klaudia Walczak
- Clinic of Neurology, Military Institute of Medicine, Warsaw, Poland,
| | - Maciej Nowocień
- Department of Radiology, Military Institute of Medicine, Warsaw, Poland
| | - Emilia Frankowska
- Department of Radiology, Military Institute of Medicine, Warsaw, Poland
| | - Adam Stępień
- Clinic of Neurology, Military Institute of Medicine, Warsaw, Poland,
| |
Collapse
|
114
|
Grover SP, Bergmeier W, Mackman N. Platelet Signaling Pathways and New Inhibitors. Arterioscler Thromb Vasc Biol 2019; 38:e28-e35. [PMID: 29563117 DOI: 10.1161/atvbaha.118.310224] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Steven P Grover
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine (S.P.G., N.M.) and McAllister Heart Institute and Department of Biochemistry and Biophysics (W.B.), University of North Carolina at Chapel Hill
| | - Wolfgang Bergmeier
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine (S.P.G., N.M.) and McAllister Heart Institute and Department of Biochemistry and Biophysics (W.B.), University of North Carolina at Chapel Hill
| | - Nigel Mackman
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine (S.P.G., N.M.) and McAllister Heart Institute and Department of Biochemistry and Biophysics (W.B.), University of North Carolina at Chapel Hill.
| |
Collapse
|
115
|
Lopez JJ, El Haouari M, Jardin I, Alonso N, Regodon S, Diez-Bello R, Redondo PC, Rosado JA. Flavonoids and Platelet-Derived Thrombotic Disorders. Curr Med Chem 2019; 26:7035-7047. [DOI: 10.2174/0929867325666180417170218] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 12/14/2017] [Accepted: 12/14/2017] [Indexed: 01/12/2023]
Abstract
:
Thrombotic disorders are characterized by an increase in the probability of the
formation of unnecessary thrombi that might be due to the activation of the coagulation cascade
or the circulating platelets. Platelets or thrombocytes play an essential role in hemostasis
but abnormal platelet function leads to the development of a number of cardiovascular
complications, including thrombotic disorders. Under pathological conditions, platelets are
associated with the development of different thrombotic disorders, including atherosclerosis,
arterial thrombosis and stroke, deep venous thrombosis and pulmonary embolism; therefore,
platelets are the target of a number of anti-thrombotic strategies. Flavonoids, a large group
of polyphenols ubiquitously expressed in fruits and vegetables that have attracted considerable
attention because of their benefits in human health, including the reduction of the risk
of cardiovascular disease. Flavonoids have been reported to reduce platelet activity by attenuating
agonist-induced GPIIb/IIIa receptor activation, mobilization of intracellular free
Ca2+, granule exocytosis, as well as activation of different signaling molecules such as mitogen-
activated protein kinases or phospholipases. This review summarizes the current studies
concerning the modulation of platelet activation by flavonoids, giving especial attention to
those events associated to thrombotic disorders.
Collapse
Affiliation(s)
- Jose J. Lopez
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003-Cáceres, Spain
| | - Mohammed El Haouari
- Faculté Polydisciplinaire de Taza, Laboratoire des Matériaux, Substances Naturelles, Environnement et Modélisation (LMSNEM), Université Sidi Mohamed Ben Abdellah, B.P. 1223, Taza Gare, Morocco
| | - Isaac Jardin
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003-Cáceres, Spain
| | - Nieves Alonso
- Department of Hematology, Hospital Infanta Cristina, 06006 Badajoz, Spain
| | - Sergio Regodon
- Department of Animal Medicine, University of Extremadura, 10003-Cáceres, Spain
| | - Raquel Diez-Bello
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003-Cáceres, Spain
| | - Pedro C. Redondo
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003-Cáceres, Spain
| | - Juan A. Rosado
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003-Cáceres, Spain
| |
Collapse
|
116
|
Rayes J, Watson SP, Nieswandt B. Functional significance of the platelet immune receptors GPVI and CLEC-2. J Clin Invest 2019; 129:12-23. [PMID: 30601137 DOI: 10.1172/jci122955] [Citation(s) in RCA: 217] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Although platelets are best known for their role in hemostasis, they are also crucial in development, host defense, inflammation, and tissue repair. Many of these roles are regulated by the immune-like receptors glycoprotein VI (GPVI) and C-type lectin receptor 2 (CLEC-2), which signal through an immunoreceptor tyrosine-based activation motif (ITAM). GPVI is activated by collagen in the subendothelial matrix, by fibrin and fibrinogen in the thrombus, and by a remarkable number of other ligands. CLEC-2 is activated by the transmembrane protein podoplanin, which is found outside of the vasculature and is upregulated in development, inflammation, and cancer, but there is also evidence for additional ligands. In this Review, we discuss the physiological and pathological roles of CLEC-2 and GPVI and their potential as targets in thrombosis and thrombo-inflammatory disorders (i.e., disorders in which inflammation plays a critical role in the ensuing thrombosis) relative to current antiplatelet drugs.
Collapse
Affiliation(s)
- Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, United Kingdom
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| |
Collapse
|
117
|
Zhang X, Zhang W, Wu X, Li H, Zhang C, Huang Z, Shi R, You T, Shi J, Cao Y. Prognostic Significance of Plasma CLEC-2 (C-Type Lectin-Like Receptor 2) in Patients With Acute Ischemic Stroke. Stroke 2019; 50:45-52. [PMID: 30580704 DOI: 10.1161/strokeaha.118.022563] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- CLEC-2 (C-type lectin-like receptor 2) is a C-type lectin receptor highly expressed on platelets with the prominent involvement in platelet activation, which was increased in coronary heart disease. Given the role of platelet activation in ischemic stroke and the connections between coronary heart disease and ischemic stroke, CLEC-2 might be a candidate marker of ischemic stroke. Here, we aimed to examine the prognostic significance of CLEC-2 in patients with acute ischemic stroke (AIS). Methods- Three hundred fifty-two patients with AIS within 7 days and 112 healthy controls were prospectively studied. Plasma CLEC-2 (pCLEC-2) and some conventional risk factors of stroke were examined. Stroke progression was defined as any new neurological symptoms/signs or any neurological worsening within 7 days after stroke onset, and poor prognosis was defined as modified Rankin Scale scores >2 at 90 days. The association between pCLEC-2 and stroke progression/prognosis was evaluated using regression models. Results- Patients with AIS had a significantly higher level of pCLEC-2 than that of healthy controls (P<0.05). Patients with AIS with progressive stroke or poor prognosis had a much higher level of pCLEC-2 compared with those with stable stroke or good prognosis (all P<0.05). Increasing pCLEC-2 was significantly associated with an increased risk of stroke progression (odds ratio, 1.97; 95% CI, 1.11-3.50; P=0.021) and poor prognosis (odds ratio, 1.70; 95% CI, 1.17-2.48; P=0.006). Patients with the highest pCLEC-2 level were 7- to 8-fold more likely to have stroke progression compared with the lowest quartile (odds ratio, 7.69; 95% CI, 1.43-41.41). Patients with the highest pCLEC-2 level were also more likely to have poor prognosis at 90 days (odds ratio, 5.58; 95% CI, 1.76-17.68). The optimal cutoff points of pCLEC-2 for predicting stroke progression and poor prognosis were 235.48 and 207.08 pg/mL, respectively. Conclusions- Increased pCLEC-2 was associated with stroke progression and poor prognosis at 90 days significantly, which indicates the prognostic role of pCLEC-2 in AIS. However, it needs to be confirmed in large-scale studies.
Collapse
Affiliation(s)
- Xia Zhang
- From the Department of Neurology (X.Z., W.Z., X.W., C.Z., Z.H., R.S., J.S., Y.C.), Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Zhang
- From the Department of Neurology (X.Z., W.Z., X.W., C.Z., Z.H., R.S., J.S., Y.C.), Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xuechun Wu
- From the Department of Neurology (X.Z., W.Z., X.W., C.Z., Z.H., R.S., J.S., Y.C.), Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hui Li
- Department of Cardiology (H.L., T.Y.), Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Chunyuan Zhang
- From the Department of Neurology (X.Z., W.Z., X.W., C.Z., Z.H., R.S., J.S., Y.C.), Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhichao Huang
- From the Department of Neurology (X.Z., W.Z., X.W., C.Z., Z.H., R.S., J.S., Y.C.), Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Rongfang Shi
- From the Department of Neurology (X.Z., W.Z., X.W., C.Z., Z.H., R.S., J.S., Y.C.), Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Tao You
- Department of Cardiology (H.L., T.Y.), Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jijun Shi
- From the Department of Neurology (X.Z., W.Z., X.W., C.Z., Z.H., R.S., J.S., Y.C.), Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yongjun Cao
- From the Department of Neurology (X.Z., W.Z., X.W., C.Z., Z.H., R.S., J.S., Y.C.), Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
118
|
Antiplatelet Drugs in the Management of Cerebral Ischemia. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00057-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
119
|
Hamdan F, Mahmood I, Al-Tameemi W. Blood cell activation as an indicator of prothrombogenesis in polycythemia vera: Case control study. IRAQI JOURNAL OF HEMATOLOGY 2019. [DOI: 10.4103/ijh.ijh_10_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
120
|
Baaten CCFMJ, Meacham S, de Witt SM, Feijge MAH, Adams DJ, Akkerman JWN, Cosemans JMEM, Grassi L, Jupe S, Kostadima M, Mattheij NJA, Prins MH, Ramirez-Solis R, Soehnlein O, Swieringa F, Weber C, White JK, Ouwehand WH, Heemskerk JWM. A synthesis approach of mouse studies to identify genes and proteins in arterial thrombosis and bleeding. Blood 2018; 132:e35-e46. [PMID: 30275110 PMCID: PMC6293874 DOI: 10.1182/blood-2018-02-831982] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 09/19/2018] [Indexed: 12/25/2022] Open
Abstract
Antithrombotic therapies reduce cardiovascular diseases by preventing arterial thrombosis and thromboembolism, but at expense of increased bleeding risks. Arterial thrombosis studies using genetically modified mice have been invaluable for identification of new molecular targets. Because of low sample sizes and heterogeneity in approaches or methodologies, a formal meta-analysis to compare studies of mice with single-gene defects encountered major limitations. To overcome these, we developed a novel synthesis approach to quantitatively scale 1514 published studies of arterial thrombus formation (in vivo and in vitro), thromboembolism, and tail-bleeding of genetically modified mice. Using a newly defined consistency parameter (CP), indicating the strength of published data, comparisons were made of 431 mouse genes, of which 17 consistently contributed to thrombus formation without affecting hemostasis. Ranking analysis indicated high correlations between collagen-dependent thrombosis models in vivo (FeCl3 injury or ligation/compression) and in vitro. Integration of scores and CP values resulted in a network of protein interactions in thrombosis and hemostasis (PITH), which was combined with databases of genetically linked human bleeding and thrombotic disorders. The network contained 2946 nodes linked to modifying genes of thrombus formation, mostly with expression in megakaryocytes. Reactome pathway analysis and network characteristics revealed multiple novel genes with potential contribution to thrombosis/hemostasis. Studies with additional knockout mice revealed that 4 of 8 (Apoe, Fpr2, Ifnar1, Vps13a) new genes were modifying in thrombus formation. The PITH network further: (i) revealed a high similarity of murine and human hemostatic and thrombotic processes and (ii) identified multiple new candidate proteins regulating these processes.
Collapse
Affiliation(s)
- Constance C F M J Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Stuart Meacham
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge, United Kingdom
| | - Susanne M de Witt
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Marion A H Feijge
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - David J Adams
- Wellcome Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Jan-Willem N Akkerman
- Laboratory of Clinical Chemistry and Haematology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Judith M E M Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Luigi Grassi
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge, United Kingdom
| | - Steve Jupe
- EMBL-European Bioinformatics Institute, Cambridge, United Kingdom
| | - Myrto Kostadima
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge, United Kingdom
| | - Nadine J A Mattheij
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Martin H Prins
- Department of Clinical Epidemiology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | | | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany; and
- Department of Pathology, AMC, Amsterdam, The Netherlands
| | - Frauke Swieringa
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University, Munich, Germany
- DZHK, Partner Site Munich Heart Alliance, Munich, Germany; and
| | | | - Willem H Ouwehand
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge, United Kingdom
- Wellcome Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
121
|
Store-operated calcium entry in thrombosis and thrombo-inflammation. Cell Calcium 2018; 77:39-48. [PMID: 30530092 DOI: 10.1016/j.ceca.2018.11.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/31/2018] [Accepted: 11/14/2018] [Indexed: 01/03/2023]
Abstract
Cytosolic free calcium (Ca2+) is a second messenger regulating a wide variety of functions in blood cells, including adhesion, activation, proliferation and migration. Store-operated Ca2+ entry (SOCE), triggered by depletion of Ca2+ from the endoplasmic reticulum, provides a main mechanism of regulated Ca2+ influx in blood cells. SOCE is mediated and regulated by isoforms of the ion channel proteins ORAI and TRP, and the transmembrane Ca2+ sensors stromal interaction molecules (STIMs), respectively. This report provides an overview of the (patho)physiological importance of SOCE in blood cells implicated in thrombosis and thrombo-inflammation, i.e. platelets and immune cells. We also discuss the physiological consequences of dysregulated SOCE in platelets and immune cells and the potential of SOCE inhibition as a therapeutic option to prevent or treat arterial thrombosis as well as thrombo-inflammatory disease states such as ischemic stroke.
Collapse
|
122
|
Eisinger F, Patzelt J, Langer HF. The Platelet Response to Tissue Injury. Front Med (Lausanne) 2018; 5:317. [PMID: 30483508 PMCID: PMC6242949 DOI: 10.3389/fmed.2018.00317] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 10/23/2018] [Indexed: 12/16/2022] Open
Abstract
In recent years, various studies have increasingly explained platelet functions not only in their central role as a regulator in cellular hemostasis and coagulation. In fact, there is growing evidence that under specific conditions, platelets act as a mediator between the vascular system, hemostasis, and the immune system. Therefore, they are essential in many processes involved in tissue remodeling and tissue reorganization after injury or inflammatory responses. These processes include the promotion of inflammatory processes, the contribution to innate and adaptive immune responses during bacterial and viral infections, the modulation of angiogenesis, and the regulation of cell apoptosis in steady-state tissue homeostasis or after tissue breakdown. All in all platelets may contribute to the control of tissue homeostasis much more than generally assumed. This review summarizes the current knowledge of platelets as part of the tissue remodeling network and seeks to provide possible translational implications for clinical therapy.
Collapse
Affiliation(s)
- Felix Eisinger
- Section for Cardioimmunology, Department of Cardiovascular Medicine, University of Tuebingen, Tübingen, Germany
| | - Johannes Patzelt
- University Clinic for Cardiovascular Medicine, University of Tuebingen, Tübingen, Germany
| | - Harald F. Langer
- Section for Cardioimmunology, Department of Cardiovascular Medicine, University of Tuebingen, Tübingen, Germany
- University Clinic for Cardiovascular Medicine, University of Tuebingen, Tübingen, Germany
| |
Collapse
|
123
|
Wei G, Luo Q, Wang X, Wu X, Xu M, Ding N, Zhao Y, Zhong L, Wang J, Wu Y, Li X, Liu Y, Ju W, Li Z, Zeng L, Xu K, Qiao J. Increased GPIbα shedding from platelets treated with immune thrombocytopenia plasma. Int Immunopharmacol 2018; 66:91-98. [PMID: 30445311 DOI: 10.1016/j.intimp.2018.11.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/08/2018] [Accepted: 11/08/2018] [Indexed: 12/19/2022]
Abstract
Immune thrombocytopenia (ITP) is a heterogeneous autoimmune disease, characterized by accelerated platelet destruction/clearance or decreased platelet production. ADAM17-mediated platelet receptor GPIbα extracellular domain shedding has been shown to be involved in platelet clearance. Whether GPIbα shedding participates in the pathogenesis of ITP remains poorly understood. This study aims to investigate the role of GPIbα shedding in the development of ITP via incubating normal platelets with ITP plasma to mimic ITP in vivo environment. Plasma was isolated from ITP patients or healthy control and incubated with platelets in vitro followed by measuring GPIbα expression by flow cytometry and western blot, ADAM17 expression by western blot, ROS generation and platelet activation by flow cytometry. Compared with control plasma, ITP plasma-treated platelet displayed significantly reduced GPIbα surface expression, increased ADAM17 expression and ROS generation. However, metalloproteinase inhibitor GM6001 blocked the ITP-plasma-induced decrease in GPIbα surface expression, increase in ADAM17 expression and platelet activation. In addition, inhibitors of NADPH oxidase or mitochondria respiration significantly inhibited ROS generation from ITP plasma-treated platelets. Moreover, ROS inhibition or blocking FcγRIIa attenuated the decrease in GPIbα surface expression, platelet activation and ROS generation (for blocking FcγRIIa) in ITP plasma-treated platelets. In conclusion, ITP plasma induces platelet receptor GPIbα extracellular domain shedding, suggesting that it might participate in the pathogenesis of ITP and targeting it might be a novel approach for treating ITP.
Collapse
Affiliation(s)
- Guangyu Wei
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Qi Luo
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Xiamin Wang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Xiaoqing Wu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, Xinyi City Hospital, Xuzhou, China
| | - Mengdi Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Ning Ding
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Yan Zhao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Lamei Zhong
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Jurui Wang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Yulu Wu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Xiaoqian Li
- Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yun Liu
- Department of Clinical Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wen Ju
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Zhenyu Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.
| |
Collapse
|
124
|
Paez-Mayorga J, Chen AL, Kotla S, Tao Y, Abe RJ, He ED, Danysh BP, Hofmann MCC, Le NT. Ponatinib Activates an Inflammatory Response in Endothelial Cells via ERK5 SUMOylation. Front Cardiovasc Med 2018; 5:125. [PMID: 30238007 PMCID: PMC6135907 DOI: 10.3389/fcvm.2018.00125] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/20/2018] [Indexed: 12/18/2022] Open
Abstract
Ponatinib is a multi-targeted third generation tyrosine kinase inhibitor (TKI) used in the treatment of chronic myeloid leukemia (CML) patients harboring the Abelson (Abl)-breakpoint cluster region (Bcr) T315I mutation. In spite of having superb clinical efficacy, ponatinib triggers severe vascular adverse events (VAEs) that significantly limit its therapeutic potential. On vascular endothelial cells (ECs), ponatinib promotes EC dysfunction and apoptosis, and inhibits angiogenesis. Furthermore, ponatinib-mediated anti-angiogenic effect has been suggested to play a partial role in systemic and pulmonary hypertension via inhibition of vascular endothelial growth factor receptor 2 (VEGFR2). Even though ponatinib-associated VAEs are well documented, their etiology remains largely unknown, making it difficult to efficiently counteract treatment-related adversities. Therefore, a better understanding of the mechanisms by which ponatinib mediates VAEs is critical. In cultured human aortic ECs (HAECs) treated with ponatinib, we found an increase in nuclear factor NF-kB/p65 phosphorylation and NF-kB activity, inflammatory gene expression, cell permeability, and cell apoptosis. Mechanistically, ponatinib abolished extracellular signal-regulated kinase 5 (ERK5) transcriptional activity even under activation by its upstream kinase mitogen-activated protein kinase kinase 5α (CA-MEK5α). Ponatinib also diminished expression of ERK5 responsive genes such as Krüppel-like Factor 2/4 (klf2/4) and eNOS. Because ERK5 SUMOylation counteracts its transcriptional activity, we examined the effect of ponatinib on ERK5 SUMOylation, and found that ERK5 SUMOylation is increased by ponatinib. We also found that ponatibib-mediated increased inflammatory gene expression and decreased anti-inflammatory gene expression were reversed when ERK5 SUMOylation was inhibited endogenously or exogenously. Overall, we propose a novel mechanism by which ponatinib up-regulates endothelial ERK5 SUMOylation and shifts ECs to an inflammatory phenotype, disrupting vascular homeostasis.
Collapse
Affiliation(s)
- Jesus Paez-Mayorga
- Department of Cardiovascular Sciences, Center of Cardiovascular Regeneration Houston, Methodist Research Institute, Methodist Hospital, Houston, TX, United States
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Mexico
| | - Andrew L. Chen
- Department of Cardiovascular Sciences, Center of Cardiovascular Regeneration Houston, Methodist Research Institute, Methodist Hospital, Houston, TX, United States
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yunting Tao
- Department of Cardiovascular Sciences, Center of Cardiovascular Regeneration Houston, Methodist Research Institute, Methodist Hospital, Houston, TX, United States
| | - Rei J. Abe
- Department of Cardiovascular Sciences, Center of Cardiovascular Regeneration Houston, Methodist Research Institute, Methodist Hospital, Houston, TX, United States
| | - Emma D. He
- Department of Cardiovascular Sciences, Center of Cardiovascular Regeneration Houston, Methodist Research Institute, Methodist Hospital, Houston, TX, United States
| | - Brian P. Danysh
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marie-Claude C. Hofmann
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Center of Cardiovascular Regeneration Houston, Methodist Research Institute, Methodist Hospital, Houston, TX, United States
| |
Collapse
|
125
|
Functional redundancy between RAP1 isoforms in murine platelet production and function. Blood 2018; 132:1951-1962. [PMID: 30131434 DOI: 10.1182/blood-2018-03-838714] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 08/11/2018] [Indexed: 01/14/2023] Open
Abstract
RAP GTPases, important regulators of cellular adhesion, are abundant signaling molecules in the platelet/megakaryocytic lineage. However, mice lacking the predominant isoform, RAP1B, display a partial platelet integrin activation defect and have a normal platelet count, suggesting the existence of a RAP1-independent pathway to integrin activation in platelets and a negligible role for RAP GTPases in megakaryocyte biology. To determine the importance of individual RAP isoforms on platelet production and on platelet activation at sites of mechanical injury or vascular leakage, we generated mice with megakaryocyte-specific deletion (mKO) of Rap1a and/or Rap1b Interestingly, Rap1a/b-mKO mice displayed a marked macrothrombocytopenia due to impaired proplatelet formation by megakaryocytes. In platelets, RAP isoforms had redundant and isoform-specific functions. Deletion of RAP1B, but not RAP1A, significantly reduced α-granule secretion and activation of the cytoskeleton regulator RAC1. Both isoforms significantly contributed to thromboxane A2 generation and the inside-out activation of platelet integrins. Combined deficiency of RAP1A and RAP1B markedly impaired platelet aggregation, spreading, and clot retraction. Consistently, thrombus formation in physiological flow conditions was abolished in Rap1a/b-mKO, but not Rap1a-mKO or Rap1b-mKO, platelets. Rap1a/b-mKO mice were strongly protected from experimental thrombosis and exhibited a severe defect in hemostasis after mechanical injury. Surprisingly, Rap1a/b-mKO platelets were indistinguishable from controls in their ability to prevent blood-lymphatic mixing during development and hemorrhage at sites of inflammation. In summary, our studies demonstrate an essential role for RAP1 signaling in platelet integrin activation and a critical role in platelet production. Although important for hemostatic/thrombotic plug formation, platelet RAP1 signaling is dispensable for vascular integrity during development and inflammation.
Collapse
|
126
|
NaveenKumar SK, SharathBabu BN, Hemshekhar M, Kemparaju K, Girish KS, Mugesh G. The Role of Reactive Oxygen Species and Ferroptosis in Heme-Mediated Activation of Human Platelets. ACS Chem Biol 2018; 13:1996-2002. [PMID: 29869870 DOI: 10.1021/acschembio.8b00458] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Hemolysis, a process by which the destruction of red blood cells leads to the release of hemoglobin, is a critical event observed during hemolytic disorders. Under oxidative stress conditions, hemoglobin can release its heme prosthetic group, which is highly cytotoxic and can catalyze the generation of reactive oxygen species (ROS), leading to several undesired redox reactions in the cells. Herein, we demonstrate for the first time that heme can mediate the activation and death of human platelets through ferroptosis, which is an iron-dependent form of nonapoptotic cell death. This study also suggests that the heme-mediated lipid peroxidation and ferroptosis in platelets may play an important role in hemolytic disorders.
Collapse
Affiliation(s)
- Somanathapura K. NaveenKumar
- DOS in Biochemistry, University of Mysore, Manasagangotri, Mysuru 570 006, India
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560 012, India
| | - Bidare N. SharathBabu
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560 012, India
| | - Mahadevappa Hemshekhar
- Department of Internal Medicine, Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg R3E3P4, Canada
| | - Kempaiah Kemparaju
- DOS in Biochemistry, University of Mysore, Manasagangotri, Mysuru 570 006, India
| | - Kesturu S. Girish
- DOS in Biochemistry, University of Mysore, Manasagangotri, Mysuru 570 006, India
- Department of Studies and Research in Biochemistry, Tumkur University, Tumakuru 572 103, India
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560 012, India
| |
Collapse
|
127
|
Burchell SR, Tang J, Zhang JH. Hematoma Expansion Following Intracerebral Hemorrhage: Mechanisms Targeting the Coagulation Cascade and Platelet Activation. Curr Drug Targets 2018; 18:1329-1344. [PMID: 28378693 DOI: 10.2174/1389450118666170329152305] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/20/2016] [Accepted: 03/14/2017] [Indexed: 01/04/2023]
Abstract
Hematoma expansion (HE), defined as a greater than 33% increase in intracerebral hemorrhage (ICH) volume within the first 24 hours, results in significant neurological deficits, and enhancement of ICH-induced primary and secondary brain injury. An escalation in the use of oral anticoagulants has led to a surge in the incidences of oral anticoagulation-associated ICH (OAT-ICH), which has been associated with a greater risk for HE and worse functional outcomes following ICH. The oral anticoagulants in use include vitamin K antagonists, and direct thrombin and factor Xa inhibitors. Fibrinolytic agents are also frequently administered. These all act via differing mechanisms and thus have varying degrees of impact on HE and ICH outcome. Additionally, antiplatelet medications have also been increasingly prescribed, and result in increased bleeding risks and worse outcomes after ICH. Aspirin, thienopyridines, and GPIIb/IIIa receptor blockers are some of the most common agents in use clinically, and also have different effects on ICH and hemorrhage growth, based on their mechanisms of action. Recent studies have found that reduced platelet activity may be more effective in predicting ICH risk, hemorrhage expansion, and outcomes, than antiplatelet agents, and activating platelets may thus be a novel target for ICH therapy. This review explores how dysfunctions or alterations in the coagulation and platelet cascades can lead to, and/or exacerbate, hematoma expansion following intracerebral hemorrhage, and describe the mechanisms behind these effects and the drugs that induce them. We also discuss potential future therapy aimed at increasing platelet activity after ICH.
Collapse
Affiliation(s)
- Sherrefa R Burchell
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda CA, USA.,Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Jiping Tang
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda CA, USA.,Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda CA, USA.,Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, USA.,Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda CA, USA
| |
Collapse
|
128
|
Zhang JX, Guo JM, Lin HJ, Zhang TT, Li ZG, Zhou JC, Zhang ZZ. Neuroprotective effects of Yiqihuoxue calm wind capsule on ischemic stroke in rats. Chin J Nat Med 2018; 15:758-765. [PMID: 29103461 DOI: 10.1016/s1875-5364(17)30107-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Indexed: 02/02/2023]
Abstract
Stroke remains the third leading cause of death and of adult disability worldwide. Vascular occlusion, followed by ischemic cascade, leads to irreversible tissue injury. Recombinant tissue plasminogen activator is the only FDA approved drug for the current treatment of acute ischemic stroke. However, traditional Chinese medicine has a long history and rich clinical experience in the treatment and rehabilitation of ischemic stroke. Using a classical middle cerebral artery occlusion (MCAO) stroke model, we tested the effectiveness of Yiqihuoxue calm wind (YCW) capsule on neurological function, gross pathology and oxidative stress status in MCAO rats. YCW capsule (3.36 and 6.72 g·kg-1 of crude drug) could significantly lower Longa's score and superoxide dismutase (SOD) level, together with less necrotic cells and infarcted area. In addition to elevated MDA and downregulated iNOS expression, YCW capsule exhibited its neuroprotective effects via free radical scavenging and NO inhibition.
Collapse
Affiliation(s)
- Jun-Xia Zhang
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China; Department of Pharmacology, Henan Provincial Institute of Food and Drug Control, Zhengzhou 450003, China
| | - Jiao-Mei Guo
- Department of Pharmacology, Henan Provincial Institute of Food and Drug Control, Zhengzhou 450003, China
| | - Hong-Jun Lin
- Department of Pharmacology, Henan Provincial Institute of Food and Drug Control, Zhengzhou 450003, China
| | - Ting-Ting Zhang
- Department of Pharmacology, Henan Provincial Institute of Food and Drug Control, Zhengzhou 450003, China
| | - Zhen-Guo Li
- Department of Pharmacology, Henan Provincial Institute of Food and Drug Control, Zhengzhou 450003, China
| | - Ji-Chun Zhou
- Department of Pharmacology, Henan Provincial Institute of Food and Drug Control, Zhengzhou 450003, China
| | - Zhen-Zhong Zhang
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases of Henan Province, Zhengzhou 450001, China.
| |
Collapse
|
129
|
|
130
|
Vögtle T, Cherpokova D, Bender M, Nieswandt B. Targeting platelet receptors in thrombotic and thrombo-inflammatory disorders. Hamostaseologie 2017; 35:235-43. [DOI: 10.5482/hamo-14-10-0049] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/21/2015] [Indexed: 12/20/2022] Open
Abstract
SummaryPlatelet activation at sites of vascular injury is critical for the formation of a hemostatic plug which limits excessive blood loss, but also represents a major pathomechanism of ischemic cardio- and cerebrovascular diseases. Although currently available antiplatelet therapies have proved beneficial in preventing the recurrence of vascular events, their adverse effects on primary hemostasis emphasize the necessity to identify and characterize novel pharmacological targets for platelet inhibition. Increasing experimental evidence has suggested that several major platelet surface receptors which regulate initial steps of platelet adhesion and activation may become promising new targets for anti-platelet drugs due to their involvement in thrombotic and thrombo-inflammatory signaling cascades.This review summarizes recent developments in understanding the function of glycoprotein (GP)Ib, GPVI and the C-type lectin-like receptor 2 (CLEC-2) in hemostasis, arterial thrombosis and thrombo-inflammation and will discuss the suitability of the receptors as novel targets to treat these diseases in humans.
Collapse
|
131
|
Lee MY, Verni CC, Herbig BA, Diamond SL. Soluble fibrin causes an acquired platelet glycoprotein VI signaling defect: implications for coagulopathy. J Thromb Haemost 2017; 15:2396-2407. [PMID: 28981200 PMCID: PMC5716900 DOI: 10.1111/jth.13863] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Indexed: 11/27/2022]
Abstract
Essentials Collagen and thrombin when used simultaneously generate highly activated platelets. The effect of thrombin stimulation on subsequent glycoprotein VI (GPVI) function was observed. Soluble fibrin, but not protease activated receptor (PAR) activation, prevented GPVI activation. Circulating soluble fibrin in coagulopathic blood may cause an acquired GPVI signaling defect. SUMMARY Background In coagulopathic blood, circulating thrombin may drive platelet dysfunction. Methods/Results Using calcium dye-loaded platelets, the effect of thrombin exposure and soluble fibrin generation on subsequent platelet GPVI function was investigated. Exposure of apixaban-treated platelet-rich plasma (12% PRP) to thrombin (1-10 nm), but not ADP or thromboxane mimetic U46619 exposure, dramatically blocked subsequent GPVI activation by convulxin, collagen-related peptide or fibrillar collagen. Consistent with soluble fibrin multimerizing and binding GPVI, the onset of convulxin insensitivity required 200-500 s of thrombin exposure, was not mimicked by exposure to PAR-1/4 activating peptides, was not observed with washed platelets, and was blocked by fibrin polymerization inhibitor (GPRP) or factor XIIIa inhibitor (T101). PAR-1 signaling through Gαq was not required because vorapaxar blocked thrombin-induced calcium mobilization but had no effect on the ability of thrombin to impair GPVI-signaling. Convulxin insensitivity was unaffected by the metalloprotease inhibitor GM6001 or the αIIb β3 antagonist GR144053, indicating negligible roles for GPVI shedding or αIIb β3 binding of fibrin. Thrombin treatment of washed platelets resuspended in purified fibrinogen also produced convulxin insensitivity that was prevented by GPRP. Exposure of apixaban/PPACK-treated whole blood to thrombin-treated fibrinogen resulted in > 50% decrease in platelet deposition in a collagen microfluidic assay that required soluble fibrin assembly. Conclusions Conversion of only 1% plasma fibrinogen in coagulopathic blood would generate 90 nm soluble fibrin, far exceeding ~1 nmGPVI in blood. Soluble fibrin, rather than thrombin-induced platelet activation throuh PAR-1 and PAR-4, downregulated GPVI-signaling in response to stimuli, and may lead to subsequent hypofunction of endogenous or transfused platelets.
Collapse
Affiliation(s)
- Mei Yan Lee
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher C. Verni
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bradley A. Herbig
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Scott L. Diamond
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
132
|
Kraft P, Schleicher R, Olbrich M, Schuhmann MK, Blanz K, Emschermann F, Ebenhoech S, Hilgendorf I, Meuth SG, Edlich F, Kleinschnitz C, Starz C, Langer HF. Platelet derived FasL contributes to apoptosis in stroke. Thromb Haemost 2017; 116:998-1000. [DOI: 10.1160/th16-06-0447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 08/07/2016] [Indexed: 01/23/2023]
|
133
|
Jamasbi J, Ayabe K, Goto S, Nieswandt B, Peter K, Siess W. Platelet receptors as therapeutic targets: Past, present and future. Thromb Haemost 2017; 117:1249-1257. [DOI: 10.1160/th16-12-0911] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 04/08/2017] [Indexed: 01/08/2023]
Abstract
SummaryAnti-platelet drugs reduce arterial thrombosis after plaque rupture and erosion, prevent stent thrombosis and are used to prevent and treat myocardial infarction and ischaemic stroke. Some of them may also be helpful in treating less frequent diseases such as thrombotic thrombocytopenic purpura. The present concise review aims to cover current and future developments of anti-platelet drugs interfering with the interaction of von Willebrand factor (VWF) with glycoprotein (GP) Ibα, and directed against GPVI, GPIIb/IIIa (integrin αIIbβ3), the thrombin receptor PAR-1, and the ADP receptor P2Y12. The high expectations of having novel antiplatelet drugs which selectively inhibit arterial thrombosis without interfering with normal haemostasis could possibly be met in the near future.
Collapse
|
134
|
Hou Y, Zhou H, Wang Y, Marshall A, Liang C, Dai X, Li BX, Vanhoorelbeke K, Lei X, Reheman A, Ni H. Anfibatide, a novel GPIb complex antagonist, inhibits platelet adhesion and thrombus formation in vitro and in vivo in murine models of thrombosis. Thromb Haemost 2017; 111:279-89. [DOI: 10.1160/th13-06-0490] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 09/25/2013] [Indexed: 01/01/2023]
Abstract
SummaryPlatelet adhesion and aggregation at the sites of vascular injury are key events for thrombosis and haemostasis. It has been well demonstrated that interaction between glycoprotein (GP) Ib and von Willebrand factor (VWF) initiates platelet adhesion and contributes to platelet aggregation, particularly at high shear. GPIb has long been suggested as a desirable antithrombotic target, but anti-GPIb therapy has never been successfully developed. Here, we evaluated the antithrombotic potential of Anfibatide, a novel snake venom-derived GPIb antagonist. We found Anfibatide inhibited washed murine platelet aggregation induced by ristocetin and recombinant murine VWF. It also blocked botrocetin-induced binding of murine plasma VWF to recombinant human GPIb . Interestingly, Anfibatide did not inhibit botrocetin- induced aggregation of platelet-rich plasma, indicating that its binding site may differ from other snake venom-derived GPIb antagonists. Anfibatide strongly inhibited platelet adhesion, aggregation, and thrombus formation in perfusion chambers at high shear conditions and efficiently dissolved preformed thrombi. Anfibatide also inhibited thrombus growth at low shear conditions, though less than at high shear. Using intravital microscopy, we found that Anfibatide markedly inhibited thrombosis in laser-injured cremaster vessels and prevented vessel occlusion in FeCl3-injured mesenteric vessels. Importantly, Anfibatide further inhibited residual thrombosis in VWF-deficient mice, suggesting that Anfibatide has additional antithrombotic effect beyond its inhibitory role in GPIb-VWF interaction. Anfibatide did not significantly cause platelet activation, prolong tail bleeding time, or cause bleeding diathesis in mice. Thus, consistent with the data from an ongoing clinical trial, the data from this study suggests that Anfibatide is a potent and safe antithrombotic agent.
Collapse
|
135
|
Łopatkiewicz G, Buda S, Mlynarski J. Application of the EF and GH Fragments to the Synthesis of Idraparinux. J Org Chem 2017; 82:12701-12714. [DOI: 10.1021/acs.joc.7b02497] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Grzegorz Łopatkiewicz
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Szymon Buda
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Jacek Mlynarski
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| |
Collapse
|
136
|
Human endoglin as a potential new partner involved in platelet-endothelium interactions. Cell Mol Life Sci 2017; 75:1269-1284. [PMID: 29080903 PMCID: PMC5843676 DOI: 10.1007/s00018-017-2694-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 10/20/2017] [Accepted: 10/24/2017] [Indexed: 11/20/2022]
Abstract
Complex interactions between platelets and activated endothelium occur during the thrombo-inflammatory reaction at sites of vascular injuries and during vascular hemostasis. The endothelial receptor endoglin is involved in inflammation through integrin-mediated leukocyte adhesion and transmigration; and heterozygous mutations in the endoglin gene cause hereditary hemorrhagic telangiectasia type 1. This vascular disease is characterized by a bleeding tendency that is postulated to be a consequence of telangiectasia fragility rather than a platelet defect, since platelets display normal functions in vitro in this condition. Here, we hypothesize that endoglin may act as an adhesion molecule involved in the interaction between endothelial cells and platelets through integrin recognition. We find that the extracellular domain of human endoglin promotes specific platelet adhesion under static conditions and confers resistance of adherent platelets to detachment upon exposure to flow. Also, platelets adhere to confluent endothelial cells in an endoglin-mediated process. Remarkably, Chinese hamster ovary cells ectopically expressing the human αIIbβ3 integrin acquire the capacity to adhere to myoblast transfectants expressing human endoglin, whereas platelets from Glanzmann’s thrombasthenia patients lacking the αIIbβ3 integrin are defective for endoglin-dependent adhesion to endothelial cells. Furthermore, the bleeding time, but not the prothrombin time, is significantly prolonged in endoglin-haplodeficient (Eng+/−) mice compared to Eng+/+ animals. These results suggest a new role for endoglin in αIIbβ3 integrin-mediated adhesion of platelets to the endothelium, and may provide a better understanding on the basic cellular mechanisms involved in hemostasis and thrombo-inflammatory events.
Collapse
|
137
|
Yao Y, Chen Y, Adili R, McKeown T, Chen P, Zhu G, Li D, Ling W, Ni H, Yang Y. Plant-based Food Cyanidin-3-Glucoside Modulates Human Platelet Glycoprotein VI Signaling and Inhibits Platelet Activation and Thrombus Formation. J Nutr 2017; 147:1917-1925. [PMID: 28855423 DOI: 10.3945/jn.116.245944] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 01/19/2017] [Accepted: 07/19/2017] [Indexed: 11/14/2022] Open
Abstract
Background: Platelets play an important role in hemostasis, thrombosis, and atherosclerosis. Glycoprotein VI (GPVI) is a major platelet receptor that interacts with exposed collagen on injured vessel walls. Our previous studies have shown that anthocyanins (a type of natural plant pigment) attenuate platelet function; however, whether anthocyanins affect collagen-induced GPVI signaling remains unknown.Objective: The objective of this study was to explore the effects of cyanidin-3-glucoside (Cy-3-g, one of the major bioactive compounds in anthocyanins) on platelet activation and thrombosis and the GPVI signaling pathway.Methods: Platelets from healthy men and women were isolated and incubated with different concentrations (0, 0.5, 5, and 50 μM) of Cy-3-g. The expression of activated integrin αIIbβ3, P-selectin, CD63, and CD40L, fibrinogen binding to platelets, and platelet aggregation were evaluated in vitro. Platelet adhesion and aggregation in whole blood under flow conditions were assessed in collagen-coated perfusion chambers. Thrombosis and hemostasis were assessed in 3-4-wk-old male C57BL/6J mice through FeCl3-induced intravital microscopy and tail bleeding time. The effect of Cy-3-g on collagen-induced human platelet GPVI signaling was explored with Western blot.Results: Cy-3-g attenuated platelet function in a dose-dependent manner. The 0.5-μM dose of Cy-3-g inhibited (P < 0.05) human platelet adhesion and aggregation to collagen at both venous (-54.02%) and arterial (-22.90%) shear stresses. The 5-μM dose inhibited (P < 0.05) collagen-induced human platelet activation (PAC-1: -48.21%, P-selectin: -50.63%), secretion (CD63: -73.89%, CD40L: -43.70%), fibrinogen binding (-56.79%), and aggregation (-17.81%). The 5-μM dose attenuated (P < 0.01) thrombus growth (-66.67%) without prolonging bleeding time in mice. The 50-μM dose downregulated (P < 0.05) collagen-induced GPVI signaling in human platelets and significantly decreased phosphorylation of Syk-linker for activation of T cells (LAT)-SLP76 (Syk: -39.08%, LAT: -32.25%, SLP76: -40.00%) and the expression of Lyn (-31.89%), Fyn (-36.27%), and phospholipase C-γ2 (-39.08%).Conclusions: Cy-3-g inhibits human platelet activation, aggregation, secretion, and thrombus formation, and downregulates the collagen-GPVI signaling pathway. Supplementation of Cy-3-g may have protective effects against atherothrombosis.
Collapse
Affiliation(s)
- Yanling Yao
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, People's Republic of China
| | - Yanqiu Chen
- Guangzhou Women and Children's Medical Centre, Guangzhou, People's Republic of China
| | - Reheman Adili
- Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Departments of
| | - Thomas McKeown
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Departments of
| | - Pingguo Chen
- Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Departments of
| | - Guangheng Zhu
- Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Departments of
| | - Dan Li
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, People's Republic of China
| | - Wenhua Ling
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, People's Republic of China
| | - Heyu Ni
- Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Ontario, Canada; .,Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada; Departments of.,Laboratory Medicine and Pathobiology.,Physiology, and.,Medicine, University of Toronto, Toronto, Ontario, Canada; and.,Canadian Blood Services, Toronto, Ontario, Canada
| | - Yan Yang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China; .,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, People's Republic of China
| |
Collapse
|
138
|
Dupuis-Girod S, Cottin V, Shovlin CL. The Lung in Hereditary Hemorrhagic Telangiectasia. Respiration 2017; 94:315-330. [PMID: 28850955 DOI: 10.1159/000479632] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is a dominantly inherited genetic vascular disorder with an estimated prevalence of 1 in 6,000, characterized by recurrent epistaxis, cutaneous telangiectasia, and arteriovenous malformations (AVMs) that affect many organs including the lungs, gastrointestinal tract, liver, and brain. Its diagnosis is based on the Curaçao criteria, and is considered definite if at least 3 of the 4 following criteria are fulfilled: (1) spontaneous and recurrent epistaxis, (2) telangiectasia, (3) a family history, and (4) pulmonary, liver, cerebral, spinal, or gastrointestinal AVMs. The focus of this review is on delineating how HHT affects the lung.
Collapse
Affiliation(s)
- Sophie Dupuis-Girod
- Service de génétique - centre de référence national pour la maladie de Rendu-Osler, Hôpital Femme-Mère-Enfants, Hospices Civils de Lyon, Bron, France
| | | | | |
Collapse
|
139
|
Bieber K, Ernst AL, Tukaj S, Holtsche MM, Schmidt E, Zillikens D, Ludwig RJ, Kasperkiewicz M. Analysis of serum markers of cellular immune activation in patients with bullous pemphigoid. Exp Dermatol 2017; 26:1248-1252. [PMID: 28500685 DOI: 10.1111/exd.13382] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2017] [Indexed: 12/21/2022]
Abstract
Experimental models of bullous pemphigoid (BP), the most frequent subepidermal autoimmune bullous disease, revealed that the immune response leading to blister formation represents an incompletely understood complex process involving different inflammatory cells. In contrast to previous reports commonly focusing on limited molecular and cellular phenotypes of the disease, the aim of this study was to investigate a broad spectrum of markers of cellular immune activation in patients with BP. We found that serum levels of soluble CD4, myeloperoxidase, S100A12, eosinophil cationic protein and soluble P-selectin were significantly elevated in patients with active BP compared with normal controls. Mast cell tryptase and neopterin serum levels significantly decreased at the time of clinical remission of the patients. Additionally, serum concentrations of soluble IL-2 receptor, mast cell tryptase and soluble P-selectin were significantly associated with levels of circulating anti-BP180 autoantibodies. Our findings confirm and extend previous reports suggesting some concomitant involvement of a panel of molecules representative for a wide spectrum of cellular players (T cells, mast cells, neutrophils, eosinophils, macrophages and platelets) orchestrating the inflammatory reaction in BP. These data may favour the employment of broad-spectrum or combined immunosuppressants, potentially together with an anticoagulant treatment, over cell- or molecule-specific targeted therapy in patients with this disorder.
Collapse
Affiliation(s)
- Katja Bieber
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Anna Lara Ernst
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Stefan Tukaj
- Department of Molecular Biology, University of Gdańsk, Gdańsk, Poland
| | - Maike M Holtsche
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Enno Schmidt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany.,Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Detlef Zillikens
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany.,Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Ralf J Ludwig
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany.,Department of Dermatology, University of Lübeck, Lübeck, Germany
| | | |
Collapse
|
140
|
Beck S, Leitges M, Stegner D. Protein kinase Cι/λ is dispensable for platelet function in thrombosis and hemostasis in mice. Cell Signal 2017; 38:223-229. [PMID: 28739484 DOI: 10.1016/j.cellsig.2017.07.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/05/2017] [Accepted: 07/20/2017] [Indexed: 11/17/2022]
Abstract
Platelet activation at sites of vascular injury is crucial for hemostasis, but it may also cause myocardial infarction or ischemic stroke. Upon platelet activation, cytoskeletal reorganization is essential for platelet secretion and thrombus formation. Members of the protein kinase C family, which includes 12 isoforms, are involved in most platelet responses required for thrombus formation. The atypical protein kinase Cι/λ (PKCι/λ) has been implicated as an important mediator of cell polarity, carcinogenesis and immune cell responses. PKCι/λ is known to be associated with the small GTPase Cdc42, an important mediator of multiple platelet functions; however, its exact function in platelets is not known. To study the role of PKCι/λ, we generated platelet- and megakaryocyte-specific PKCι/λ knockout mice (Prkcifl/fl, Pf4-Cre) and used them to investigate the function of PKCι/λ in platelet activation and aggregation in vitro and in vivo. Surprisingly, lack of PKCι/λ had no detectable effect on platelet spreading and function in vitro and in vivo under all tested conditions. These results indicate that PKCι/λ is dispensable for Cdc42-triggered processes and for thrombosis and hemostasis in mice.
Collapse
Affiliation(s)
- Sarah Beck
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | | | - David Stegner
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
141
|
Lien LM, Lin KH, Huang LT, Tseng MF, Chiu HC, Chen RJ, Lu WJ. Licochalcone A Prevents Platelet Activation and Thrombus Formation through the Inhibition of PLCγ2-PKC, Akt, and MAPK Pathways. Int J Mol Sci 2017; 18:ijms18071500. [PMID: 28704925 PMCID: PMC5535990 DOI: 10.3390/ijms18071500] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/07/2017] [Accepted: 07/09/2017] [Indexed: 01/27/2023] Open
Abstract
Platelet activation is involved in cardiovascular diseases, such as atherosclerosis and ischemic stroke. Licochalcone A (LA), an active ingredient of licorice, exhibits multiple biological activities such as anti-oxidation and anti-inflammation. However, its role in platelet activation remains unclear. Therefore, the study investigated the antiplatelet mechanism of LA. Our data revealed that LA (2-10 μM) concentration dependently inhibited platelet aggregation induced by collagen, but not thrombin and U46619. LA markedly attenuated collagen-stimulated ATP release, P-selectin secretion, calcium mobilization, and GPIIbIIIa activation, but did not interfere with the collagen binding to platelets. Moreover, LA significantly reduced the activation of PLCγ2, PKC, Akt and MAPKs. Thus, LA attenuates platelet activation, possibly by inhibiting collagen receptor downstream signaling but not by blocking the collagen receptors. In addition, LA prevented adenosine diphosphate (ADP)-induced acute pulmonary thrombosis, fluorescein sodium-induced platelet thrombus formation, and middle cerebral artery occlusion/reperfusion-induced brain injury in mice, but did not affect normal hemostasis. This study demonstrated that LA effectively reduced platelet activation and thrombus formation, in part, through the inhibition of PLCγ2-PKC, Akt, and MAPK pathways, without the side effect of bleeding. These findings also indicate that LA may provide a safe and alternative therapeutic approach for preventing thromboembolic disorders such as stroke.
Collapse
Affiliation(s)
- Li-Ming Lien
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Neurology, Shin Kong Wu Ho Su Memorial Hospital, Taipei 111, Taiwan.
| | - Kuan-Hung Lin
- Department of Pharmacology and Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 110, Taiwan.
- Central Laboratory, Shin Kong Wu Ho Su Memorial Hospital, Taipei 111, Taiwan.
| | - Li-Ting Huang
- Department of Medical Research and Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan.
| | - Mei-Fang Tseng
- Department of Medical Research and Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan.
| | - Hou-Chang Chiu
- Department of Neurology, Shin Kong Wu Ho Su Memorial Hospital, Taipei 111, Taiwan.
- College of Medicine, Fu-Jen Catholic University, Taipei 242, Taiwan.
| | - Ray-Jade Chen
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Medical Research and Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan.
| | - Wan-Jung Lu
- Department of Pharmacology and Graduate Institute of Medical Sciences, Taipei Medical University, Taipei 110, Taiwan.
- Department of Medical Research and Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan.
- Graduate Institute of Metabolism and Obesity Sciences, College of Public Health and Nutrition, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
142
|
Lebozec K, Jandrot-Perrus M, Avenard G, Favre-Bulle O, Billiald P. Design, development and characterization of ACT017, a humanized Fab that blocks platelet's glycoprotein VI function without causing bleeding risks. MAbs 2017; 9:945-958. [PMID: 28598281 PMCID: PMC5540112 DOI: 10.1080/19420862.2017.1336592] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Glycoprotein VI is a platelet-specific collagen receptor critical for in vivo formation of arterial thrombosis. It is also considered as an attractive target for the development of anti-thrombotic drugs because blocking glycoprotein (GP)VI inhibits platelet aggregation without inducing detrimental effects on physiologic hemostasis. Here, we present data on the identification, in vitro and ex vivo pharmacology of a humanized Fab fragment designated as ACT017. ACT017 was selected out of 15 humanized variants based upon structural and functional properties. It was produced under GMP-like conditions followed by detailed physico-chemical analysis and functional characterization indicating high antigen-binding specificity and affinity. In addition, we demonstrate, in a dose-escalation study, that ACT017 has a high capacity to specifically inhibit collagen-induced platelet aggregation ex vivo after injection to the macaque without inducing thrombocytopenia, GPVI depletion or bleeding side effects as is the case for conventional anti-platelets. Therefore, ACT017 is a promising therapeutic candidate for the development of a new generation of safe and efficient anti-thrombotic drugs.
Collapse
Affiliation(s)
- Kristell Lebozec
- a Acticor Biotech SAS, Hôpital Bichat - Inserm U1148 , 46 rue Henri Huchard, F75018 Paris , France
| | - Martine Jandrot-Perrus
- a Acticor Biotech SAS, Hôpital Bichat - Inserm U1148 , 46 rue Henri Huchard, F75018 Paris , France.,b Inserm-University Paris Diderot UMR S1148, Hôpital Bichat , 46 rue Henri Huchard, F75018 Paris , France
| | - Gilles Avenard
- a Acticor Biotech SAS, Hôpital Bichat - Inserm U1148 , 46 rue Henri Huchard, F75018 Paris , France
| | - Olivier Favre-Bulle
- a Acticor Biotech SAS, Hôpital Bichat - Inserm U1148 , 46 rue Henri Huchard, F75018 Paris , France.,c 3Biotech , 4 place Louis Armand, F75012 Paris , France
| | - Philippe Billiald
- a Acticor Biotech SAS, Hôpital Bichat - Inserm U1148 , 46 rue Henri Huchard, F75018 Paris , France.,d University Paris-Sud, University Paris-Saclay , School of Pharmacy, IPSIT , 5 rue J.-B. Clément, F92296 Châtenay-Malabry , France
| |
Collapse
|
143
|
Abstract
AbstractCVD is the leading cause of death worldwide, a consequence of mostly poor lifestyle and dietary behaviours. Although whole fruit and vegetable consumption has been consistently shown to reduce CVD risk, the exact protective constituents of these foods are yet to be clearly identified. A recent and biologically plausible hypothesis supporting the cardioprotective effects of vegetables has been linked to their inorganic nitrate content. Approximately 60–80 % inorganic nitrate exposure in the human diet is contributed by vegetable consumption. Although inorganic nitrate is a relatively stable molecule, under specific conditions it can be metabolised in the body to produce NO via the newly discovered nitrate–nitrite–NO pathway. NO is a major signalling molecule in the human body, and has a key role in maintaining vascular tone, smooth muscle cell proliferation, platelet activity and inflammation. Currently, there is accumulating evidence demonstrating that inorganic nitrate can lead to lower blood pressure and improved vascular compliance in humans. The aim of this review is to present an informative, balanced and critical review of the current evidence investigating the role of inorganic nitrate and nitrite in the development, prevention and/or treatment of CVD. Although there is evidence supporting short-term inorganic nitrate intakes for reduced blood pressure, there is a severe lack of research examining the role of long-term nitrate intakes in the treatment and/or prevention of hard CVD outcomes, such as myocardial infarction and cardiovascular mortality. Epidemiological evidence is needed in this field to justify continued research efforts.
Collapse
|
144
|
Jiun-Yi L, Ting-Chen C, Nen-Chung C, Jayakumar T, Chao-Chien C. Anti-embolic effect of Taorenchengqi Tang in rats with embolic stroke induced by occluding middle cerebral artery. J TRADIT CHIN MED 2017. [DOI: 10.1016/s0254-6272(17)30068-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
145
|
Xu S, Zhong A, Ma H, Li D, Hu Y, Xu Y, Zhang J. Neuroprotective effect of salvianolic acid B against cerebral ischemic injury in rats via the CD40/NF-κB pathway associated with suppression of platelets activation and neuroinflammation. Brain Res 2017; 1661:37-48. [DOI: 10.1016/j.brainres.2017.02.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 02/07/2017] [Accepted: 02/12/2017] [Indexed: 11/16/2022]
|
146
|
Liu X, Niu Y, Chen KC, Chen S. Rapid hemostatic and mild polyurethane-urea foam wound dressing for promoting wound healing. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 71:289-297. [DOI: 10.1016/j.msec.2016.10.019] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 09/24/2016] [Accepted: 10/14/2016] [Indexed: 10/20/2022]
|
147
|
Pleines I, Woods J, Chappaz S, Kew V, Foad N, Ballester-Beltrán J, Aurbach K, Lincetto C, Lane RM, Schevzov G, Alexander WS, Hilton DJ, Astle WJ, Downes K, Nurden P, Westbury SK, Mumford AD, Obaji SG, Collins PW, Delerue F, Ittner LM, Bryce NS, Holliday M, Lucas CA, Hardeman EC, Ouwehand WH, Gunning PW, Turro E, Tijssen MR, Kile BT. Mutations in tropomyosin 4 underlie a rare form of human macrothrombocytopenia. J Clin Invest 2017; 127:814-829. [PMID: 28134622 PMCID: PMC5330761 DOI: 10.1172/jci86154] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 12/01/2016] [Indexed: 01/12/2023] Open
Abstract
Platelets are anuclear cells that are essential for blood clotting. They are produced by large polyploid precursor cells called megakaryocytes. Previous genome-wide association studies in nearly 70,000 individuals indicated that single nucleotide variants (SNVs) in the gene encoding the actin cytoskeletal regulator tropomyosin 4 (TPM4) exert an effect on the count and volume of platelets. Platelet number and volume are independent risk factors for heart attack and stroke. Here, we have identified 2 unrelated families in the BRIDGE Bleeding and Platelet Disorders (BPD) collection who carry a TPM4 variant that causes truncation of the TPM4 protein and segregates with macrothrombocytopenia, a disorder characterized by low platelet count. N-Ethyl-N-nitrosourea–induced (ENU-induced) missense mutations in Tpm4 or targeted inactivation of the Tpm4 locus led to gene dosage–dependent macrothrombocytopenia in mice. All other blood cell counts in Tpm4-deficient mice were normal. Insufficient TPM4 expression in human and mouse megakaryocytes resulted in a defect in the terminal stages of platelet production and had a mild effect on platelet function. Together, our findings demonstrate a nonredundant role for TPM4 in platelet biogenesis in humans and mice and reveal that truncating variants in TPM4 cause a previously undescribed dominant Mendelian platelet disorder.
Collapse
Affiliation(s)
- Irina Pleines
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Joanne Woods
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Stephane Chappaz
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Verity Kew
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Nicola Foad
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - José Ballester-Beltrán
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Katja Aurbach
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Wuerzburg, Wuerzburg, Germany
| | - Chiara Lincetto
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Rachael M. Lane
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Galina Schevzov
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Warren S. Alexander
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Douglas J. Hilton
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - William J. Astle
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Kate Downes
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Paquita Nurden
- Institut Hospitalo-Universitaire LIRYC, Plateforme Technologique d’Innovation Biomédicale, Hôpital Xavier Arnozan, Pessac, France
| | - Sarah K. Westbury
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Andrew D. Mumford
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Samya G. Obaji
- Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Heath Park, Cardiff, United Kingdom
| | - Peter W. Collins
- Arthur Bloom Haemophilia Centre, School of Medicine, Cardiff University, Heath Park, Cardiff, United Kingdom
| | - NIHR BioResource
- NIHR BioResource–Rare Diseases, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Fabien Delerue
- Transgenic Animal Unit, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, Australia
| | - Lars M. Ittner
- Transgenic Animal Unit, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, Australia
| | - Nicole S. Bryce
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Mira Holliday
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Christine A. Lucas
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Edna C. Hardeman
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Willem H. Ouwehand
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NIHR BioResource–Rare Diseases, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Human Genetics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Peter W. Gunning
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Ernest Turro
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Medical Research Council Biostatistics Unit, Cambridge Institute of Public Health, Cambridge, United Kingdom
| | - Marloes R. Tijssen
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Benjamin T. Kile
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| |
Collapse
|
148
|
Alteration of platelet GPVI signaling in ST-elevation myocardial infarction patients demonstrated by a combination of proteomic, biochemical, and functional approaches. Sci Rep 2016; 6:39603. [PMID: 28004756 PMCID: PMC5177944 DOI: 10.1038/srep39603] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 11/24/2016] [Indexed: 11/30/2022] Open
Abstract
The platelet-specific collagen receptor glycoprotein VI (GPVI) is critical for the formation of arterial thrombosis in vivo. We analyzed GPVI-activated platelets from ST-elevation myocardial infarction (STEMI) patients and matched stable coronary artery disease (SCAD) controls in order to provide novel clues on the degree of involvement of GPVI signaling in the acute event. Firstly, platelets were isolated from systemic venous blood and activated with the GPVI specific agonist CRP (collagen-related peptide). STEMI and SCAD samples were compared by a phosphoproteomics approach. Validations were by immunoblotting in systemic and intracoronary blood from independent cohorts of patients. Twenty-six differentially regulated proteins were identified when comparing CRP-activated systemic platelets from STEMI and SCAD patients, 4 of which were selected for validation studies: PLCɣ2, G6f, SLP-76, and Dok-2. Immunoblot analyses showed these four proteins had higher tyrosine phosphorylation levels in response to CRP in platelets from STEMI patients, being these levels more pronounced at the culprit site of coronary artery occlusion. Moreover, platelet aggregation studies showed a higher response to GPVI agonists in STEMI patients compared to SCAD controls. In conclusion, we show an altered activation state of GPVI signaling in STEMI patients, confirming this receptor as a promising anti-thrombotic target for myocardial infarction.
Collapse
|
149
|
Timosaponin AIII induces antiplatelet and antithrombotic activity via Gq-mediated signaling by the thromboxane A2 receptor. Sci Rep 2016; 6:38757. [PMID: 27934923 PMCID: PMC5146924 DOI: 10.1038/srep38757] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 11/14/2016] [Indexed: 12/02/2022] Open
Abstract
The thromboxane (Tx) A2 pathway is a major contributor to the amplification of initial platelet activation and is therefore a key drug target. To identify potent small-molecule inhibitors of the thromboxane prostaglandin (TP) receptor, we screened a small steroidal saponin library using U46619-induced rat platelet aggregation assays. Timosaponin AIII (TAIII) was identified as a potent inhibitor of U46619-induced rat platelet aggregation and exhibited superior selectivity for the TP receptor versus other G protein-coupled receptors and a PKC activator. TAIII inhibited U46619-induced rat platelet aggregation independent of increases in cAMP and cGMP and the inhibition of TxA2 production. Both PKC and PLC activators restored TAIII-inhibited platelet aggregation, whereas TAIII did not inhibit platelet aggregation induced by co-activation of the G12/13 and Gz pathways. Furthermore, TAIII did not affect the platelet shape change or ROCK2 phosphorylation evoked by low-dose U46619. In vivo, TAIII prolonged tail bleeding time, reduced the mortality of animals with acute pulmonary thromboembolism and significantly reduced venous thrombus weight. Our study suggests that TAIII, by preferentially targeting Gq-mediated PLC/PKC signaling from the TP receptor, induces stronger in vitro antiplatelet activity and in vivo antithrombotic effects and may be an excellent candidate for the treatment of thrombotic disorders.
Collapse
|
150
|
Abstract
Ischemic disorders, such as myocardial infarction, stroke, and peripheral vascular disease, are the most common causes of debilitating disease and death in westernized cultures. The extent of tissue injury relates directly to the extent of blood flow reduction and to the length of the ischemic period, which influence the levels to which cellular ATP and intracellular pH are reduced. By impairing ATPase-dependent ion transport, ischemia causes intracellular and mitochondrial calcium levels to increase (calcium overload). Cell volume regulatory mechanisms are also disrupted by the lack of ATP, which can induce lysis of organelle and plasma membranes. Reperfusion, although required to salvage oxygen-starved tissues, produces paradoxical tissue responses that fuel the production of reactive oxygen species (oxygen paradox), sequestration of proinflammatory immunocytes in ischemic tissues, endoplasmic reticulum stress, and development of postischemic capillary no-reflow, which amplify tissue injury. These pathologic events culminate in opening of mitochondrial permeability transition pores as a common end-effector of ischemia/reperfusion (I/R)-induced cell lysis and death. Emerging concepts include the influence of the intestinal microbiome, fetal programming, epigenetic changes, and microparticles in the pathogenesis of I/R. The overall goal of this review is to describe these and other mechanisms that contribute to I/R injury. Because so many different deleterious events participate in I/R, it is clear that therapeutic approaches will be effective only when multiple pathologic processes are targeted. In addition, the translational significance of I/R research will be enhanced by much wider use of animal models that incorporate the complicating effects of risk factors for cardiovascular disease. © 2017 American Physiological Society. Compr Physiol 7:113-170, 2017.
Collapse
Affiliation(s)
- Theodore Kalogeris
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Christopher P. Baines
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Ronald J. Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|