51
|
Rivera J, Cid J. Editorial. Platelets 2022; 33:3-4. [PMID: 35083955 DOI: 10.1080/09537104.2021.2010952] [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]
Affiliation(s)
- José Rivera
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, Murcia
| | - Joan Cid
- Unidad de Aféresis y Terapia Celular, Servicio de Hemoterapia y Hemostasia, Institut Clínic de Malalties Hematològiques i Oncològiques (ICMHO), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universidad de Barcelona, Hospital Clínic, Barcelona, España
| |
Collapse
|
52
|
McKenzie AJ, Doyle BJ, Aman ZM. Micromechanical Force Measurement of Clotted Blood Particle Cohesion: Understanding Thromboembolic Aggregation Mechanisms. Cardiovasc Eng Technol 2022; 13:816-828. [PMID: 35419664 PMCID: PMC9750917 DOI: 10.1007/s13239-022-00618-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 03/19/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE Arterial shear forces may promote the embolization of clotted blood from the surface of thrombi, displacing particles that may occlude vasculature, with increased risk of physiological complications and mortality. Thromboemboli may also collide in vivo to form metastable aggregates that increase vessel occlusion likelihood. METHODS A micromechanical force (MMF) apparatus was modified for aqueous applications to study clot-liquid interfacial phenomena between clotted porcine blood particles suspended in modified continuous phases. The MMF measurement is based on visual observation of particle-particle separation, where Hooke's Law is applied to calculate separation force. This technique has previously been deployed to study solid-fluid interfacial phenomena in oil and gas pipelines, providing fundamental insight to cohesive and adhesive properties between solids in multiphase flow systems. RESULTS This manuscript introduces distributed inter-particle separation force properties as a function of governing physio-chemical parameters; pre-load (contact) force, contact time, and bulk phase chemical modification. In each experimental campaign, the hysteresis and distributed force properties were analysed, to derive insight as to the governing mechanism of cohesion between particles. Porcine serum, porcine albumin and pharmaceutical agents (alteplase, tranexamic acid and hydrolysed aspirin) reduced the measurement by an order of magnitude from the baseline measurement-the apparatus provides a platform to study how surface-active chemistries impact the solid-fluid interface. CONCLUSION These results provide new insight to potential mechanisms of macroscopic thromboembolic aggregation via particles cohering in the vascular system-data that can be directly applied to computational simulations to predict particle fate, better informing the mechanistic developments of embolic occlusion.
Collapse
Affiliation(s)
- Angus J. McKenzie
- grid.1012.20000 0004 1936 7910Department of Chemical Engineering, The Centre for Long Subsea Tiebacks, Fluid Science and Resources Cluster, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009 Australia
| | - Barry J. Doyle
- grid.1012.20000 0004 1936 7910Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, Nedlands, and Centre for Medical Research, The University of Western Australia, Crawley, PER Australia ,Australian Research Council Centre for Personalised Therapeutics Technologies, Parkville, Australia ,grid.4305.20000 0004 1936 7988BHF Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
| | - Zachary M. Aman
- grid.1012.20000 0004 1936 7910Department of Chemical Engineering, The Centre for Long Subsea Tiebacks, Fluid Science and Resources Cluster, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009 Australia
| |
Collapse
|
53
|
Regulation of Key Antiplatelet Pathways by Bioactive Compounds with Minimal Bleeding Risk. Int J Mol Sci 2021; 22:ijms222212380. [PMID: 34830261 PMCID: PMC8620148 DOI: 10.3390/ijms222212380] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 01/04/2023] Open
Abstract
Cardiovascular disease is strongly influenced by platelet activation. Platelet activation and thrombus formation at atherosclerotic plaque rupture sites is a dynamic process regulated by different signaling networks. Therefore, there are now focused efforts to search for novel bioactive compounds which target receptors and pathways in the platelet activation process while preserving normal hemostatic function. The antiplatelet activity of numerous fruits and vegetables and their multiple mechanisms of action have recently been highlighted. In this review, we review the antiplatelet actions of bioactive compounds via key pathways (protein disulfide isomerase, mitogen-activated protein kinases, mitochondrial function, cyclic adenosine monophosphate, Akt, and shear stress-induced platelet aggregation) with no effects on bleeding time. Therefore, targeting these pathways might lead to the development of effective antiplatelet strategies that do not increase the risk of bleeding.
Collapse
|
54
|
Multiparameter Evaluation of the Platelet-Inhibitory Effects of Tyrosine Kinase Inhibitors Used for Cancer Treatment. Int J Mol Sci 2021; 22:ijms222011199. [PMID: 34681859 PMCID: PMC8540269 DOI: 10.3390/ijms222011199] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/01/2021] [Accepted: 10/13/2021] [Indexed: 12/30/2022] Open
Abstract
Current antiplatelet drugs for the treatment of arterial thrombosis often coincide with increased bleeding risk. Several tyrosine kinase inhibitors (TKIs) for cancer treatment inhibit platelet function, with minor reported bleeding symptoms. The aim of this study was to compare the antiplatelet properties of eight TKIs to explore their possible repurposing as antiplatelet drugs. Samples of whole blood, platelet-rich plasma (PRP), or isolated platelets from healthy donors were treated with TKI or the vehicle. Measurements of platelet aggregation, activation, intracellular calcium mobilization, and whole-blood thrombus formation under flow were performed. Dasatinib and sunitinib dose-dependently reduced collagen-induced aggregation in PRP and washed platelets; pazopanib, cabozantinib, and vatalanib inhibited this response in washed platelets only; and fostamatinib, axitinib, and lapatinib showed no/limited effects. Fostamatinib reduced thrombus formation by approximately 50% on collagen and other substrates. Pazopanib, sunitinib, dasatinib, axitinib, and vatalanib mildly reduced thrombus formation on collagen by 10–50%. Intracellular calcium responses in isolated platelets were inhibited by dasatinib (>90%), fostamatinib (57%), sunitinib (77%), and pazopanib (82%). Upon glycoprotein-VI receptor stimulation, fostamatinib, cabozantinib, and vatalanib decreased highly activated platelet populations by approximately 15%, while increasing resting populations by 39%. In conclusion, the TKIs with the highest affinities for platelet-expressed molecular targets most strongly inhibited platelet functions. Dasatinib, fostamatinib, sunitinib, and pazopanib interfered in early collagen receptor-induced molecular-signaling compared with cabozantinib and vatalanib. Fostamatinib, sunitinib, pazopanib, and vatalanib may be promising for future evaluation as antiplatelet drugs.
Collapse
|
55
|
Ferrer-Raventós P, Beyer K. Alternative platelet activation pathways and their role in neurodegenerative diseases. Neurobiol Dis 2021; 159:105512. [PMID: 34537329 DOI: 10.1016/j.nbd.2021.105512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 12/24/2022] Open
Abstract
PURPOSE OF THE REVIEW The study of platelets in the context of neurodegenerative diseases is intensifying, and increasing evidence suggests that platelets may play an important role in the pathogenesis of neurodegenerative disorders. Therefore, we aim to provide a comprehensive overview of the role of platelets and their diverse activation pathways in the development of these diseases. RECENT FINDINGS Platelets participate in synaptic plasticity, learning, memory, and platelets activated by exercise promote neuronal differentiation in several brain regions. Platelets also contribute to the immune response by modulating their surface protein profile and releasing pro- and anti-inflammatory mediators. In Alzheimer's disease, increased levels of platelet amyloid precursor protein raise the production of amyloid-beta peptides promoting platelet activation, triggering at the same time amyloid-beta fibrillation. In Parkinson's disease, increased platelet α-synuclein is associated with elevated ROS production and mitochondrial dysfunction. SUMMARY In this review, we revise different platelet activation pathways, those classically involved in hemostasis and wound healing, and alternative activation pathways recently described in the context of neurodegenerative diseases, especially in Alzheimer's disease.
Collapse
Affiliation(s)
- Paula Ferrer-Raventós
- Memory Unit, Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Katrin Beyer
- Department of Pathology, Germans Trias i Pujol Research Institute (IGTP), Universitat Autònoma de Barcelona (UAB), 08916 Badalona, Barcelona, Spain.
| |
Collapse
|
56
|
Al Subayyil A, Basmaeil YS, Alenzi R, Khatlani T. Human Placental Mesenchymal Stem/Stromal cells (pMSCs) inhibit agonist-induced platelet functions reducing atherosclerosis and thrombosis phenotypes. J Cell Mol Med 2021; 25:9268-9280. [PMID: 34535958 PMCID: PMC8500971 DOI: 10.1111/jcmm.16848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem/stromal cells isolated from human term placenta (pMSCs) have potential to treat clinically manifested inflammatory diseases. Atherosclerosis is a chronic inflammatory disease, and platelets play a contributory role towards its pathogenesis. During transplantation, MSCs interact with platelets and exert influence on their functional outcome. In this study, we investigated the consequences of interaction between pMSCs and platelets, and its impact on platelet-mediated atherosclerosis in vitro. Human platelets were treated with various types of pMSCs either directly or with their secretome, and their effect on agonist-mediated platelet activation and functional characteristics were evaluated. Human umbilical vein endothelial cells (HUVECs) were used as control. The impact of pMSCs treatment on platelets was evaluated by the expression of activation markers and by platelet functional analysis. A subset of pMSCs reduced agonist-induced activation of platelets, both via direct contact and with secretome treatments. Decrease in platelet activation translated into diminished spreading, limited adhesion and minimized aggregation. In addition, pMSCs decreased oxidized LDL (ox-LDL)-inducedCD36-mediated platelet activation, establishing their protective role in atherosclerosis. Gene expression and protein analysis show that pMSCs express pro- and anti-thrombotic proteins, which might be responsible for the modulation of agonist-induced platelet functions. These data suggest the therapeutic benefits of pMSCs in atherosclerosis.
Collapse
Affiliation(s)
- Abdullah Al Subayyil
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Yasser S Basmaeil
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Reem Alenzi
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Tanvir Khatlani
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
57
|
Mao S, Sarkar A, Wang Y, Song C, LeVine D, Wang X, Que L. Microfluidic chip grafted with integrin tension sensors for evaluating the effects of flowing shear stress and ROCK inhibitor on platelets. LAB ON A CHIP 2021; 21:3128-3136. [PMID: 34180491 PMCID: PMC8353964 DOI: 10.1039/d1lc00259g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/17/2021] [Indexed: 06/13/2023]
Abstract
Integrins are key players in platelet adhesion and aggregation. Integrin molecular tensions, the forces transmitted by integrin molecules, are regulated by both mechanical and biochemical cues, and the outside-in and inside-out signaling has been extensively studied. While the mechanical properties of platelets at static status have been studied by atomic force microscopy, traction force microscopy and tension sensors, the biomechanical properties of flowing platelets remain elusive. Herein, we report microfluidic chips grafted with integrin tension sensors for microfluidic-force mapping in platelets. Specifically, the process of integrin αIIbβ3 mediating tension transmission and platelet adhesion under low flow rates has been obtained, and the process of platelet clustering at post-stenotic regions has been demonstrated. We found that flowing shear force can postpone the integrin-mediated tension transmission and platelet adhesion. We further evaluated the effect of Y-27632, a ROCK inhibitor that has been proven to reduce integrin-mediated platelet adhesion, at a series of concentrations and demonstrated that microfluidic chips with integrin tension sensors are sensitive to the concentration-dependent effects of Y-27632. Given their low cost and scalable throughput, these chips are ideal technical platforms for biological studies of platelets at flowing status and for platelet inhibitor or potential antiplatelet drug screening.
Collapse
Affiliation(s)
- Subin Mao
- Electrical and Computer Engineering Department, Iowa State University, Ames, USA50011.
| | - Anwesha Sarkar
- Electrical and Computer Engineering Department, Iowa State University, Ames, USA50011. and Department of Physics and Astronomy, Iowa State University, Ames, USA50011.
| | - Yongliang Wang
- Department of Physics and Astronomy, Iowa State University, Ames, USA50011.
| | - Chao Song
- Electrical and Computer Engineering Department, Iowa State University, Ames, USA50011.
| | - Dana LeVine
- Veterinary Clinical Sciences, Iowa State University, Ames, USA50011
| | - Xuefeng Wang
- Department of Physics and Astronomy, Iowa State University, Ames, USA50011.
| | - Long Que
- Electrical and Computer Engineering Department, Iowa State University, Ames, USA50011.
| |
Collapse
|
58
|
Ye T, Zhang X, Li G, Wang S. Biomechanics in thrombus formation from direct cellular simulations. Phys Rev E 2021; 102:042410. [PMID: 33212741 DOI: 10.1103/physreve.102.042410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/02/2020] [Indexed: 11/07/2022]
Abstract
Numerically reproducing the process of thrombus formation is highly desired for understanding its mechanism but still remains challenging due to the polydisperse feature of blood components and their multiple biochemical or biomechanical behaviors involved. We numerically implemented a simplified version of the process from the perspective of biomechanics, using a mesoscale particle-based method, smoothed dissipative particle dynamics-immersed boundary method. This version covers the adhesion and aggregation of platelets (PLTs), the deformation and aggregation of red blood cells (RBCs), and the interaction between PLTs and RBCs, as well as the blockage of microvessels. Four critical factors that can affect thrombus formation were investigated: the velocity of blood flow, the adhesive ability of PLTs, the interaction strength between PLTs and RBCs, and the deformability of RBCs. Increasing the velocity of blood flow was found to be the most effective way to reduce the microvessel blockage, and reducing the adhesive ability of PLTs is also a direct and efficient way. However, decreasing the interaction strength between PLTs and RBCs sometimes does not alleviate thrombus formation, and similarly, increasing the deformability of RBCs does not have a significant improvement for the severely blocked microvessel. These results imply that maintaining high-rate blood flow plays a crucial role in the prevention and treatment of thrombosis, which is even more effective than antiplatelet or anticoagulant drugs. The drugs or treatments concentrating on reducing the PLT-RBC interaction or softening the RBCs may not have a significant effect on the thrombosis.
Collapse
Affiliation(s)
- Ting Ye
- Department of Computational Mathematics, School of Mathematics, Jilin University, Changchun, 130012, China
| | - Xuejiao Zhang
- Department of Computational Mathematics, School of Mathematics, Jilin University, Changchun, 130012, China
| | - Guansheng Li
- Department of Computational Mathematics, School of Mathematics, Jilin University, Changchun, 130012, China
| | - Sitong Wang
- Department of Computational Mathematics, School of Mathematics, Jilin University, Changchun, 130012, China
| |
Collapse
|
59
|
Mencarini T, Roka-Moiia Y, Bozzi S, Redaelli A, Slepian MJ. Electrical impedance vs. light transmission aggregometry: Testing platelet reactivity to antiplatelet drugs using the MICELI POC impedance aggregometer as compared to a commercial predecessor. Thromb Res 2021; 204:66-75. [PMID: 34147831 PMCID: PMC11416791 DOI: 10.1016/j.thromres.2021.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/08/2021] [Accepted: 05/26/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND Patients' responses to antiplatelet therapy significantly vary, with individuals showing high residual platelet reactivity associated with thrombosis. To personalize thrombosis management, platelet function testing has been suggested as a promising tool able to monitor the antithrombotic effect of antiplatelet agents in real-time. We have prototyped the MICELI, a miniature and easy-to-use electrical impedance aggregometer (EIA), measuring platelet aggregation in whole blood. Here, we tested the capability of the MICELI aggregometer to quantify platelet reactivity on antiplatelet agents, as compared with conventional light-transmission aggregometry (LTA). METHODS Platelet aggregation in ACD-anticoagulated whole blood and platelet-rich plasma of healthy donors (n = 30) was evaluated. The effect of clopidogrel, ticagrelor, cangrelor, cilostazol, and tirofiban on ADP-induced aggregation was tested, while aspirin was evaluated with arachidonic acid and collagen. Platelet aggregation was recorded using the MICELI or BioData PAP-8E (Bio/Data Corp.) aggregometers. RESULTS The MICELI aggregometer detected an adequate and comparable dose-dependent decrease of platelet aggregation in response to increments of drugs' concentrations, as compared to LTA (the inter-device R2 = 0.79-0.93). Platelet aggregation in platelet-rich plasma recorded by LTA showed higher sensitivity to antiplatelet agents, but it couldn't distinguish between different drug doses as indicated by saturation of the aggregatory response. CONCLUSION Platelet aggregation in whole blood as recorded by EIA represents a better model system for evaluation of platelet reactivity as compared with platelet aggregation in platelet-rich plasma as recorded by LTA, since EIA takes into consideration the modulatory effect of other blood cells on platelet hemostatic function and pharmacodynamics of antiplatelet drugs in vivo. As such, the MICELI impedance aggregometer could be potentially employed for the point-of-care monitoring of platelet function in patients on-treatment for personalized tailoring of their antiplatelet regimen.
Collapse
Affiliation(s)
- Tatiana Mencarini
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Yana Roka-Moiia
- Department of Medicine, Sarver Heart Center, University of Arizona, Tucson, AZ, United States of America; Department of Biomedical Engineering, Sarver Heart Center, University of Arizona, Tucson, AZ, United States of America
| | - Silvia Bozzi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Alberto Redaelli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Marvin J Slepian
- Department of Medicine, Sarver Heart Center, University of Arizona, Tucson, AZ, United States of America; Department of Biomedical Engineering, Sarver Heart Center, University of Arizona, Tucson, AZ, United States of America.
| |
Collapse
|
60
|
Zhang S, Gui X, Ding Y, Tong H, Ju W, Li Y, Li Z, Zeng L, Xu K, Qiao J. Matrine Impairs Platelet Function and Thrombosis and Inhibits ROS Production. Front Pharmacol 2021; 12:717725. [PMID: 34366869 PMCID: PMC8339414 DOI: 10.3389/fphar.2021.717725] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/15/2021] [Indexed: 12/13/2022] Open
Abstract
Matrine is a naturally occurring alkaloid and possesses a wide range of pharmacological properties, such as anti-cancer, anti-oxidant, anti-inflammatory effects. However, whether it affects platelet function and thrombosis remains unclear. This study aims to evaluate the effect of matrine on platelet function and thrombus formation. Human platelets were treated with matrine (0–1 mg/ml) for 1 h at 37°C followed by measuring platelet aggregation, granule secretion, receptor expression by flow cytometry, spreading and clot retraction. In addition, matrine (10 mg/kg) was injected intraperitoneally into mice to measure tail bleeding time, arterial and venous thrombus formation. Matrine dose-dependently inhibited platelet aggregation and ATP release in response to either collagen-related peptide (Collagen-related peptide, 0.1 μg/ml) or thrombin (0.04 U/mL) stimulation without altering the expression of P-selectin, glycoprotein Ibα, GPVI, or αIIbβ3. In addition, matrine-treated platelets presented significantly decreased spreading on fibrinogen or collagen and clot retraction along with reduced phosphorylation of c-Src. Moreover, matrine administration significantly impaired the in vivo hemostatic function of platelets, arterial and venous thrombus formation. Furthermore, in platelets stimulated with CRP or thrombin, matrine significantly reduced Reactive oxygen species generation, inhibited the phosphorylation level of ERK1/2 (Thr202/Tyr204), p38 (Thr180/Tyr182) and AKT (Thr308/Ser473) as well as increased VASP phosphorylation (Ser239) and intracellular cGMP level. In conclusion, matrine inhibits platelet function, arterial and venous thrombosis, possibly involving inhibition of ROS generation, suggesting that matrine might be used as an antiplatelet agent for treating thrombotic or cardiovascular diseases.
Collapse
Affiliation(s)
- Sixuan Zhang
- 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, Xuzhou, China
| | - Xiang Gui
- 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, Xuzhou, China
| | - Yangyang Ding
- 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, Xuzhou, China
| | - Huan Tong
- 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, 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, Xuzhou, China
| | - Yue Li
- School of Medical Technology, Xuzhou Medical University, 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, Xuzhou, China
| | - Lingyu Zeng
- 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, Xuzhou, China.,School of Medical Technology, Xuzhou Medical University, 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, 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, Xuzhou, China
| |
Collapse
|
61
|
Andreeva T, Komsa-Penkova R, Langari A, Krumova S, Golemanov G, Georgieva GB, Taneva SG, Giosheva I, Mihaylova N, Tchorbanov A, Todinova S. Morphometric and Nanomechanical Features of Platelets from Women with Early Pregnancy Loss Provide New Evidence of the Impact of Inherited Thrombophilia. Int J Mol Sci 2021; 22:ijms22157778. [PMID: 34360543 PMCID: PMC8346153 DOI: 10.3390/ijms22157778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/16/2021] [Accepted: 07/18/2021] [Indexed: 11/25/2022] Open
Abstract
Pregnancy is associated with hypercoagulation states and increased thrombotic risk, especially in women with thrombophilia. We combine atomic force microscopy (AFM) and flow cytometry to examine the morphology and nanomechanics of platelets derived from women with early pregnancy loss (EPL) and control pregnant (CP) and non-pregnant (CNP) women. Both control groups exhibit similar morphometric parameters (height and surface roughness) and membrane stiffness of platelets. EPL patients’ platelets, on the other hand, are more activated than the control groups, with prominent cytoskeletal rearrangement. In particular, reduced membrane roughness (22.9 ± 6 nm vs. 39.1 ± 8 nm) (p < 0.05) and height (692 ± 128 nm vs. 1090 ± 131 nm) (p < 0.05), strong alteration in the membrane Young modulus, increased production of platelets’ microparticles, and higher expression of procoagulant surface markers, as well as increased occurrence of thrombophilia (FVL, FII20210A, PLA1/A2, MTHFR C677T or 4G/5G PAI-1) polymorphisms were found. We suggest that the carriage of thrombophilic mutations triggers structural and nanomechanical abnormalities in platelets, resulting in their increased activation. The activation state of platelets can be well characterized by AFM, and the morphometric and nanomechanical characteristics might serve as a new criterion for evaluation of the cause of miscarriage and offer the prospect of an innovative approach serving for diagnostic purposes.
Collapse
Affiliation(s)
- Tonya Andreeva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. Georgi Bonchev, Str. Bl. 21, 1113 Sofia, Bulgaria; (T.A.); (A.L.); (S.K.); (S.G.T.); (I.G.)
| | - Regina Komsa-Penkova
- Department of Biochemistry, Medical University, 1 St. Kliment Ohridski Str., 5800 Pleven, Bulgaria; (R.K.-P.); (G.G.); (G.B.G.)
| | - Ariana Langari
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. Georgi Bonchev, Str. Bl. 21, 1113 Sofia, Bulgaria; (T.A.); (A.L.); (S.K.); (S.G.T.); (I.G.)
| | - Sashka Krumova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. Georgi Bonchev, Str. Bl. 21, 1113 Sofia, Bulgaria; (T.A.); (A.L.); (S.K.); (S.G.T.); (I.G.)
| | - Georgi Golemanov
- Department of Biochemistry, Medical University, 1 St. Kliment Ohridski Str., 5800 Pleven, Bulgaria; (R.K.-P.); (G.G.); (G.B.G.)
| | - Galya B. Georgieva
- Department of Biochemistry, Medical University, 1 St. Kliment Ohridski Str., 5800 Pleven, Bulgaria; (R.K.-P.); (G.G.); (G.B.G.)
| | - Stefka G. Taneva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. Georgi Bonchev, Str. Bl. 21, 1113 Sofia, Bulgaria; (T.A.); (A.L.); (S.K.); (S.G.T.); (I.G.)
| | - Ina Giosheva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. Georgi Bonchev, Str. Bl. 21, 1113 Sofia, Bulgaria; (T.A.); (A.L.); (S.K.); (S.G.T.); (I.G.)
- University Obstetrics and Gynecology Hospital “Maichin Dom”, 2 Zdrave Str., 1463 Sofia, Bulgaria
| | - Nikolina Mihaylova
- Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Acad. Georgi Bonchev, Str. Bl. 26, 1113 Sofia, Bulgaria; (N.M.); (A.T.)
| | - Andrey Tchorbanov
- Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Acad. Georgi Bonchev, Str. Bl. 26, 1113 Sofia, Bulgaria; (N.M.); (A.T.)
| | - Svetla Todinova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. Georgi Bonchev, Str. Bl. 21, 1113 Sofia, Bulgaria; (T.A.); (A.L.); (S.K.); (S.G.T.); (I.G.)
- Correspondence:
| |
Collapse
|
62
|
Jain N, Corken AL, Kumar A, Davis CL, Ware J, Arthur JM. Role of Platelets in Chronic Kidney Disease. J Am Soc Nephrol 2021; 32:1551-1558. [PMID: 34140394 PMCID: PMC8425650 DOI: 10.1681/asn.2020121806] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 02/04/2023] Open
Abstract
Platelet-dependent mechanisms for excessive clotting and bleeding in CKD remain undefined. Moreover, platelets' contribution to inflammation, and specifically to CKD, are equally elusive. To date, descriptions of changes in the functional properties of circulating platelets during CKD have provided confusing interpretations. Experimental approaches that can advance our understanding of platelet dysfunction in CKD are needed, and studies that provide mechanistic insights into the dynamic relationships between thrombosis, bleeding, and inflammation associated with CKD will be essential to improve clinical management and outcomes for this vulnerable population. This article summarizes existing literature characterizing platelets in CKD and identifies areas that need further investigation.
Collapse
Affiliation(s)
- Nishank Jain
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas,Medicine Service, Central Arkansas Veterans Affairs Medical Center, Little Rock, Arkansas
| | - Adam L. Corken
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Amudha Kumar
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Clayton L. Davis
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Jerry Ware
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - John M. Arthur
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas,Medicine Service, Central Arkansas Veterans Affairs Medical Center, Little Rock, Arkansas
| |
Collapse
|
63
|
Severe Trauma and Hemorrhage Leads to Platelet Dysfunction and Changes in Cyclic Nucleotides in The Rat. Shock 2021; 53:468-475. [PMID: 31090681 DOI: 10.1097/shk.0000000000001379] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Rats subjected to polytrauma and hemorrhage develop a coagulopathy that is similar to acute coagulopathy of trauma in humans, and is associated with a rise in prothrombin time and a fall in clot strength. Because platelet aggregation accounts for a major proportion of clot strength, we set out to characterize the effects of polytrauma on platelet function. METHODS Sprague-Dawley rats were anesthetized with isoflurane. Polytrauma included laparotomy and damage to 10 cm of the small intestines, right and medial liver lobes, right leg skeletal muscle, femur fracture, and hemorrhage (40% of blood volume). No resuscitation was given. Blood samples were taken before and after trauma for the measurement of impedance electrode aggregometry, and intracellular levels of cyclic adenosine and guanosine monophosphate (cAMP, cGMP), inositol trisphosphate (IP3), and adenosine and guanosine triphosphates (ATP, GTP). RESULTS Polytrauma significantly increased the response of collagen (24%) and thrombin (12%) to stimulate platelet aggregation. However, aggregation to adenosine diphosphate (ADP) or arachidonic acid (AA) was significantly decreased at 2 (52% and 46%, respectively) and 4 h (45% and 39%). Polytrauma and hemorrhage also led to a significant early rise in cAMP (101 ± 11 to 202 ± 29 pg/mL per 1,000 platelets), mirrored by a decrease in cGMP (7.8 ± 0.9 to 0.6 ± 0.5). In addition, there was a late fall in ATP (8.1 ± 0.7 to 2.2 ± 0.6 ng/mL per 1,000 platelets) and GTP (1.5 ± 0.2 to 0.3 ± 0.1). IP3 rose initially, and then fell back to baseline. CONCLUSIONS Polytrauma and hemorrhage led to a deficit in the platelet aggregation response to ADP and AA after trauma, likely due to the early rise in cAMP, and a later fall in energy substrates, and may explain the decrease in clot strength and impaired hemostasis observed after severe trauma.
Collapse
|
64
|
Lerche CJ, Schwartz F, Theut M, Fosbøl EL, Iversen K, Bundgaard H, Høiby N, Moser C. Anti-biofilm Approach in Infective Endocarditis Exposes New Treatment Strategies for Improved Outcome. Front Cell Dev Biol 2021; 9:643335. [PMID: 34222225 PMCID: PMC8249808 DOI: 10.3389/fcell.2021.643335] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/04/2021] [Indexed: 12/13/2022] Open
Abstract
Infective endocarditis (IE) is a life-threatening infective disease with increasing incidence worldwide. From early on, in the antibiotic era, it was recognized that high-dose and long-term antibiotic therapy was correlated to improved outcome. In addition, for several of the common microbial IE etiologies, the use of combination antibiotic therapy further improves outcome. IE vegetations on affected heart valves from patients and experimental animal models resemble biofilm infections. Besides the recalcitrant nature of IE, the microorganisms often present in an aggregated form, and gradients of bacterial activity in the vegetations can be observed. Even after appropriate antibiotic therapy, such microbial formations can often be identified in surgically removed, infected heart valves. Therefore, persistent or recurrent cases of IE, after apparent initial infection control, can be related to biofilm formation in the heart valve vegetations. On this background, the present review will describe potentially novel non-antibiotic, antimicrobial approaches in IE, with special focus on anti-thrombotic strategies and hyperbaric oxygen therapy targeting the biofilm formation of the infected heart valves caused by Staphylococcus aureus. The format is translational from preclinical models to actual clinical treatment strategies.
Collapse
Affiliation(s)
- Christian Johann Lerche
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Franziska Schwartz
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Marie Theut
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Emil Loldrup Fosbøl
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kasper Iversen
- Department of Cardiology, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
- Department of Emergency Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
| | - Henning Bundgaard
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Niels Høiby
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Claus Moser
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
65
|
Bose RJ, Ha K, McCarthy JR. Bio-inspired nanomaterials as novel options for the treatment of cardiovascular disease. Drug Discov Today 2021; 26:1200-1211. [PMID: 33561512 PMCID: PMC8205945 DOI: 10.1016/j.drudis.2021.01.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 01/11/2021] [Accepted: 01/20/2021] [Indexed: 11/28/2022]
Abstract
Cardiovascular disease (CVD) and its sequelae have long been the leading causes of death and disability in the developed world. Although mortality associated with CVD has been decreasing, due in large part to novel therapeutic options, the rate of decrease has flattened. Thus, there is a great need to investigate alternate therapeutic strategies that can increase efficacy while decreasing adverse effects. Nanomaterials have been widely investigated and have emerged as promising tools for both therapeutic and diagnostic purposes in oncology; however, the potential of nanomaterials has not been extensively explored for cardiovascular medicine. In this review, we focus on recent developments in the field of nanomedicines targeted for CVDs, with a special emphasis on cell membrane-coated nanoparticles (NPs) and their applications.
Collapse
Affiliation(s)
- Rajendran Jc Bose
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, Utica, NY, USA
| | - Khan Ha
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, Utica, NY, USA
| | - Jason R McCarthy
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, Utica, NY, USA.
| |
Collapse
|
66
|
Inherited Platelet Disorders: An Updated Overview. Int J Mol Sci 2021; 22:ijms22094521. [PMID: 33926054 PMCID: PMC8123627 DOI: 10.3390/ijms22094521] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/17/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Platelets play a major role in hemostasis as ppwell as in many other physiological and pathological processes. Accordingly, production of about 1011 platelet per day as well as appropriate survival and functions are life essential events. Inherited platelet disorders (IPDs), affecting either platelet count or platelet functions, comprise a heterogenous group of about sixty rare diseases caused by molecular anomalies in many culprit genes. Their clinical relevance is highly variable according to the specific disease and even within the same type, ranging from almost negligible to life-threatening. Mucocutaneous bleeding diathesis (epistaxis, gum bleeding, purpura, menorrhagia), but also multisystemic disorders and/or malignancy comprise the clinical spectrum of IPDs. The early and accurate diagnosis of IPDs and a close patient medical follow-up is of great importance. A genotype-phenotype relationship in many IPDs makes a molecular diagnosis especially relevant to proper clinical management. Genetic diagnosis of IPDs has been greatly facilitated by the introduction of high throughput sequencing (HTS) techniques into mainstream investigation practice in these diseases. However, there are still unsolved ethical concerns on general genetic investigations. Patients should be informed and comprehend the potential implications of their genetic analysis. Unlike the progress in diagnosis, there have been no major advances in the clinical management of IPDs. Educational and preventive measures, few hemostatic drugs, platelet transfusions, thrombopoietin receptor agonists, and in life-threatening IPDs, allogeneic hematopoietic stem cell transplantation are therapeutic possibilities. Gene therapy may be a future option. Regular follow-up by a specialized hematology service with multidisciplinary support especially for syndromic IPDs is mandatory.
Collapse
|
67
|
Chun JM, Lee AY, Nam JY, Lim KS, Choe MS, Lee MY, Kim C, Kim JS. Effects of Dipsacus asperoides Extract on Monosodium Iodoacetate-Induced Osteoarthritis in Rats Based on Gene Expression Profiling. Front Pharmacol 2021; 12:615157. [PMID: 33927614 PMCID: PMC8076797 DOI: 10.3389/fphar.2021.615157] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 02/02/2021] [Indexed: 12/24/2022] Open
Abstract
The root of Dipsacus asperoides C. Y. Cheng et T. M. Ai is traditionally used as an analgesic and anti-inflammatory agent to treat pain, rheumatoid arthritis, and bone fractures. However, neither its effects on osteoarthritis (OA) nor its effects on the arthritic cartilage tissue transcriptome have not been fully investigated. In this study, we used a rat model of monosodium iodoacetate- (MIA-) induced OA to investigate the therapeutic effects of a Dipsacus asperoides ethanolic extract (DAE, 200 mg/kg for 21 days). The study first assessed joint diameter, micro-CT scans, and histopathological analysis and then conducted gene expression profiling using RNA sequencing in articular cartilage tissue. We found that DAE treatment ameliorates OA disease phenotypes; it reduced the knee joint diameter and prevented changes in the structural and histological features of the joint, thereby showing that DAE has a protective effect against OA. Based on the results of gene expression profiling and subsequent pathway analysis, we found that several canonical pathways were linked to DAE treatment, including WNT/β-catenin signaling. Taken together, the present results suggest molecular mechanism, involving gene expression changes, by which DAE has a protective effect in a rat model of MIA-induced OA.
Collapse
Affiliation(s)
- Jin Mi Chun
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju-si, Korea
| | - A Yeong Lee
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju-si, Korea
| | - Jae Yong Nam
- Bioinformatics Group, R&D Center, Insilicogen Corporation, Yongin, Korea
| | - Kyung Seob Lim
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si, Korea
| | - Mu Seog Choe
- Department of Molecular Physiology, College of Pharmacy, Kyungpook National University, Daegu, Korea
| | - Min Young Lee
- Department of Molecular Physiology, College of Pharmacy, Kyungpook National University, Daegu, Korea
| | - Chul Kim
- Korea Future Medicine Division, Korea Institute of Oriental Medicine, Daejeon, Korea
| | - Joong-Sun Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju-si, Korea
| |
Collapse
|
68
|
Garcia A, Dunoyer-Geindre S, Fish RJ, Neerman-Arbez M, Reny JL, Fontana P. Methods to Investigate miRNA Function: Focus on Platelet Reactivity. Thromb Haemost 2021; 121:409-421. [PMID: 33124028 PMCID: PMC8263142 DOI: 10.1055/s-0040-1718730] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs modulating protein production. They are key players in regulation of cell function and are considered as biomarkers in several diseases. The identification of the proteins they regulate, and their impact on cell physiology, may delineate their role as diagnostic or prognostic markers and identify new therapeutic strategies. During the last 3 decades, development of a large panel of techniques has given rise to multiple models dedicated to the study of miRNAs. Since plasma samples are easily accessible, circulating miRNAs can be studied in clinical trials. To quantify miRNAs in numerous plasma samples, the choice of extraction and purification techniques, as well as normalization procedures, are important for comparisons of miRNA levels in populations and over time. Recent advances in bioinformatics provide tools to identify putative miRNAs targets that can then be validated with dedicated assays. In vitro and in vivo approaches aim to functionally validate candidate miRNAs from correlations and to understand their impact on cellular processes. This review describes the advantages and pitfalls of the available techniques for translational research to study miRNAs with a focus on their role in regulating platelet reactivity.
Collapse
Affiliation(s)
- Alix Garcia
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - Richard J. Fish
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Marguerite Neerman-Arbez
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
- iGE3, Institute of Genetics and Genomics in Geneva, Geneva, Switzerland
| | - Jean-Luc Reny
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of General Internal Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Pierre Fontana
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Angiology and Haemostasis, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
69
|
Santos-Gallego CG, Badimon J. Overview of Aspirin and Platelet Biology. Am J Cardiol 2021; 144 Suppl 1:S2-S9. [PMID: 33706986 DOI: 10.1016/j.amjcard.2020.12.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 12/11/2020] [Indexed: 12/31/2022]
Abstract
Aspirin (ASA) has historically been one of the most important drugs in cardiology and has long been the cornerstone of antiplatelet therapy. Although its role in acute coronary syndrome remains undisputed, emerging data suggest that reappraisal of the efficacy of long-term ASA in some primary and secondary prevention may be warranted. The aim of this review is to place these new results in the context of previous evidence on aspirin by appraising the current body of evidence on its use of for cardiovascular diseases. This overview first summarizes the history of the discovery of aspirin, as well as its pharmacology and the concept of ASA resistance. We subsequently recapitulate the evidence of ASA on primary prevention and secondary prevention starting from the classical studies in order to serve as an introductory background to the examination of the most recent clinical trials that will be performed in the rest of the articles of this Supplement. Although the benefit of ASA in acute coronary syndrome remains incontrovertible, emerging evidence challenge the universal need for primary prevention, or for lifelong treatment in secondary prevention or all adults with stable coronary disease who are at highest risk for ASA-induced bleeding. The role of aspirin is quickly changing in recent times and this review provides a review for the clinician about the current role of this drug in cardiovascular care.
Collapse
|
70
|
Palma-Barqueros V, Crescente M, de la Morena ME, Chan MV, Almarza E, Revilla N, Bohdan N, Miñano A, Padilla J, Allan HE, Maffucci T, Edin ML, Zeldin DC, Mesa-Nuñez C, Damian C, Marín-Quilez A, Benito R, Martínez-Martínez I, Bermejo N, Casas-Aviles I, Alen AR, González-Porras JR, Hernández-Rivas JM, Vicente V, Corral J, Lozano ML, Warner TD, Bastida JM, Rivera J. A novel genetic variant in PTGS1 affects N-glycosylation of cyclooxygenase-1 causing a dominant-negative effect on platelet function and bleeding diathesis. Am J Hematol 2021; 96:E83-E88. [PMID: 33326144 PMCID: PMC10938055 DOI: 10.1002/ajh.26076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Verónica Palma-Barqueros
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, Murcia, Spain
| | - Marilena Crescente
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University London, 4 Newark Street, London, E1 2AT, United Kingdom
| | - María Eugenia de la Morena
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, Murcia, Spain
| | - Melissa V Chan
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University London, 4 Newark Street, London, E1 2AT, United Kingdom
- The National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, USA
| | - Elena Almarza
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT)/Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) and Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM). Madrid, Spain
| | - Nuria Revilla
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, Murcia, Spain
| | - Natalia Bohdan
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, Murcia, Spain
| | - Antonia Miñano
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, Murcia, Spain
| | - José Padilla
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, Murcia, Spain
| | - Harriet E Allan
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University London, 4 Newark Street, London, E1 2AT, United Kingdom
| | - Tania Maffucci
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Matthew L. Edin
- National Institutes of Health, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Darryl. C. Zeldin
- National Institutes of Health, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Cristina Mesa-Nuñez
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT)/Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) and Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM). Madrid, Spain
| | - Carlos Damian
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT)/Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) and Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM). Madrid, Spain
| | - Ana Marín-Quilez
- IBSAL, IBMCC, CIC, Universidad de Salamanca-CSIC, Salamanca, Spain
| | - Rocío Benito
- IBSAL, IBMCC, CIC, Universidad de Salamanca-CSIC, Salamanca, Spain
| | - Irene Martínez-Martínez
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, Murcia, Spain
| | - Nuria Bermejo
- Servicio de Hematología, Hospital San Pedro de Alcántara, Complejo Universitario de Cáceres, Cáceres, Spain
| | - Ignacio Casas-Aviles
- Servicio de Hematología, Hospital San Pedro de Alcántara, Complejo Universitario de Cáceres, Cáceres, Spain
| | - Agustín Rodríguez Alen
- Servicio de Hematología y Hemoterapia, Hospital Virgen de la Salud, Complejo Hospitalario de Toledo, Spain
| | | | - Jesús María Hernández-Rivas
- IBSAL, IBMCC, CIC, Universidad de Salamanca-CSIC, Salamanca, Spain
- Departamento de Hematología, Hospital Universitario de Salamanca, Salamanca-IBSAL
| | - Vicente Vicente
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, Murcia, Spain
| | - Javier Corral
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, Murcia, Spain
| | - María Luisa Lozano
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, Murcia, Spain
| | - Timothy D. Warner
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University London, 4 Newark Street, London, E1 2AT, United Kingdom
| | - José María Bastida
- Departamento de Hematología, Hospital Universitario de Salamanca, Salamanca-IBSAL
- On behalf of the “Grupo Español de Alteraciones Plquetarias Congénitas, (GEAPC)”; Hemorrhagic Diathesis Working Group, SETH
| | - José Rivera
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, Murcia, Spain
- On behalf of the “Grupo Español de Alteraciones Plquetarias Congénitas, (GEAPC)”; Hemorrhagic Diathesis Working Group, SETH
| |
Collapse
|
71
|
Yuan J, Jiang Z, Li M, Li W, Gu X, Wang Z, Pi L, Xu Y, Zhou H, Zhang B, Deng Q, Wang Y, Huang P, Zhang L, Gu X. Integrin α2 gene polymorphism is a risk factor of coronary artery lesions in Chinese children with Kawasaki disease. Pediatr Rheumatol Online J 2021; 19:12. [PMID: 33557870 PMCID: PMC7869497 DOI: 10.1186/s12969-021-00494-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 01/11/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Kawasaki disease (KD) is a systemic vasculitis, and the formation of coronary artery lesions(CAL) is its most common sequela. Both genetic and environmental factors are considered to be important factors of in KD. Integrin α2 (ITGA2) is a transmembrane receptor that is associated with susceptibility to several diseases, but its relevance to KD with CAL is unclear. METHODS We genotyped ITGA2 rs1126643 in 785 KD patients with the CAL and no-CAL(NCAL) (300 patients with CAL, and 485 age- and sex-matched patients with NCAL). OR (95% CI) and adjusted OR (95% CI) were used to evaluate the intensity of the association. RESULTS We found a significantly increased risk of KD with CAL associated with ITGA2 rs1126643 genotypes (CT vs CC: adjusted OR = 1.57, 95% CI = 1.16-2.12, P = 0.0032; CT/TT vs CC: adjusted OR = 1.49, 95% CI = 1.12-2.00, P = 0.0068; T vs C: adjusted OR = 1.66, 95% CI = 1.16-2.51, P = 0.0165). Moreover, we found that carriers of the CT/TT genotype had a significant risk of KD with coronary artery lesion susceptibility for children ≤60 months of age, and the CT/TT genotype was significantly associated with an increased risk of SCAL formation and MCAL formation when compared with the CC genotype. CONCLUSION ITGA2 rs1126643 was associated with increased susceptibility and severity of CAL in KD.
Collapse
Affiliation(s)
- Jia Yuan
- grid.410737.60000 0000 8653 1072Department of Cardiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Zhiyong Jiang
- grid.410737.60000 0000 8653 1072Department of Blood Transfusion and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Meiai Li
- grid.410737.60000 0000 8653 1072Department of Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Wei Li
- grid.410737.60000 0000 8653 1072Department of Cardiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Xueping Gu
- grid.410737.60000 0000 8653 1072Department of Blood Transfusion and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Zhouping Wang
- grid.410737.60000 0000 8653 1072Department of Cardiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Lei Pi
- grid.410737.60000 0000 8653 1072Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Yufen Xu
- grid.410737.60000 0000 8653 1072Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Huazhong Zhou
- grid.410737.60000 0000 8653 1072Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Baidu Zhang
- grid.410737.60000 0000 8653 1072Department of Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Qiulian Deng
- grid.410737.60000 0000 8653 1072Department of Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Yanfei Wang
- grid.410737.60000 0000 8653 1072Department of Cardiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Ping Huang
- grid.410737.60000 0000 8653 1072Department of Cardiology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Li Zhang
- Department of Cardiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China.
| | - Xiaoqiong Gu
- Department of Blood Transfusion , Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China.
| |
Collapse
|
72
|
Wang W, Yang L, Song L, Guo M, Li C, Yang B, Wang M, Kou N, Gao J, Qu H, Ma Y, Xue M, Shi D. Combination of Panax notoginseng saponins and aspirin potentiates platelet inhibition with alleviated gastric injury via modulating arachidonic acid metabolism. Biomed Pharmacother 2021; 134:111165. [PMID: 33370633 DOI: 10.1016/j.biopha.2020.111165] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
High platelet reactivity and gastric mucosal injury after aspirin (ASA) treatment are associated with poor compliance and an increased risk of cardiovascular events. Panax notoginseng saponins (PNS) have been widely used for the treatment of coronary heart disease (CHD) in addition to antiplatelet drugs in China; however, the joint effect and possible mechanism of PNS in addition to ASA on platelet activation and gastric injury remain unclear. This study was designed to investigate the combinational effects of PNS with ASA, and to explore the underlying mechanism via arachidonic acid (AA) metabolism pathway using lipidomic analysis. In a randomized, assessor-blinded trial, 42 patients with stable coronary heart disease (SCHD) and chronic gastritis were randomly assigned to receive ASA (n = 21) or PNS + ASA (n = 21) for 2 months. Compared with ASA alone, PNS + ASA further inhibited CD62p expression, GPIIb-IIIa activation and platelet aggregation and led to increased platelet inhibition rate. PNS + ASA suppressed the activity of platelet cyclooxygenase (COX)-1, and decreased the production of TXB2, PGD2, PGE2, 11-HETE, the downstream oxylipids of AA/COX-1 pathway in platelets, compared with ASA alone. The severity of dyspepsia assessment (SODA) results showed that patients in PNS + ASA group exhibited relieved dyspeptic symptoms as compared with those in ASA group, which might be associated with enhanced secretion of gastrin and motilin. In vivo study of myocardial infarction rats demonstrated that PNS attenuated ASA-induced gastric mucosal injury, which was related to markedly boosted gastric level of 6,15-diketo-13,14-dihydro-prostaglandin (PG)F1α, 13,14-dihydro-15-keto-PGE2 and PGE2 from AA/PG pathway in response to PNS + ASA compared with ASA alone. In summary, our study demonstrated that the combination of PNS and ASA potentiated the antiplatelet effect of ASA via AA/COX-1/TXB2 pathway in platelets, and mitigated ASA-related gastric injury via AA/PG pathway in gastric mucosa.
Collapse
Affiliation(s)
- Wenting Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; Hangzhou Red Cross Hospital, Hangzhou 310003, China
| | - Lin Yang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Lei Song
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Ming Guo
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Changkun Li
- Shimadzu (China) Co., LTD Beijing Branch, Beijing 100020, China
| | - Bin Yang
- Department of Pathology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Mingming Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Na Kou
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Jie Gao
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Hua Qu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yan Ma
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology & Immunology, Vienna General Hospital, Medical University of Vienna, 1090, Vienna, Austria
| | - Mei Xue
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| | - Dazhuo Shi
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| |
Collapse
|
73
|
Zheng Y, Montague SJ, Lim YJ, Xu T, Xu T, Gardiner EE, Lee WM. Label-free multimodal quantitative imaging flow assay for intrathrombus formation in vitro. Biophys J 2021; 120:791-804. [PMID: 33513336 DOI: 10.1016/j.bpj.2021.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/17/2020] [Accepted: 01/13/2021] [Indexed: 10/22/2022] Open
Abstract
Microfluidics in vitro assays recapitulate a blood vessel microenvironment using surface-immobilized agonists under biofluidic flows. However, these assays do not quantify intrathrombus mass and activities of adhesive platelets at the agonist margin and use fluorescence labeling, therefore limiting clinical translation potential. Here, we describe a label-free multimodal quantitative imaging flow assay that combines rotating optical coherent scattering microscopy and quantitative phase microscopy. The combined imaging platform enables real-time evaluation of membrane fluctuations of adhesive-only platelets and total intrathrombus mass under physiological flow rates in vitro. We call this multimodal quantitative imaging flow assay coherent optical scattering and phase interferometry (COSI). COSI records intrathrombus mass to picogram accuracy and shape changes to a platelet membrane with high spatial-temporal resolution (0.4 μm/s) under physiological and pathophysiological fluid shear stress (1800 and 7500 s-1). With COSI, we generate an axial slice of 4 μm from the coverslip surface, approximately equivalent to the thickness of a single platelet, which permits nanoscale quantification of membrane fluctuation (activity) of adhesive platelets during initial adhesion, spreading, and recruitment into a developing thrombus (mass). Under fluid shear, pretreatment with a broad range metalloproteinase inhibitor (250 μM GM6001) blocked shedding of platelet adhesion receptors that shown elevated adhesive platelet activity at average of 42.1 μm/s and minimal change in intrathrombus mass.
Collapse
Affiliation(s)
- Yujie Zheng
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research
| | - Samantha J Montague
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research
| | - Yean J Lim
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research; ACRF Centre for Intravital Imaging of Niches for Cancer Immune Therapy
| | - Tao Xu
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research
| | - Tienan Xu
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research
| | - Elizabeth E Gardiner
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research
| | - Woei Ming Lee
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research; ACRF Centre for Intravital Imaging of Niches for Cancer Immune Therapy; The ARC Centre of Excellence in Advanced Molecular Imaging, The Australian National University, Canberra, Australia.
| |
Collapse
|
74
|
Spurgeon BEJ, Michelson AD, Frelinger AL. Platelet mass cytometry: Optimization of sample, reagent, and analysis parameters. Cytometry A 2021; 99:170-179. [DOI: 10.1002/cyto.a.24300] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/27/2020] [Accepted: 12/23/2020] [Indexed: 12/28/2022]
Affiliation(s)
- Benjamin E. J. Spurgeon
- Center for Platelet Research Studies, Dana‐Farber/Boston Children's Cancer and Blood Disorders Center Harvard Medical School Boston Massachusetts USA
| | - Alan D. Michelson
- Center for Platelet Research Studies, Dana‐Farber/Boston Children's Cancer and Blood Disorders Center Harvard Medical School Boston Massachusetts USA
| | - Andrew L. Frelinger
- Center for Platelet Research Studies, Dana‐Farber/Boston Children's Cancer and Blood Disorders Center Harvard Medical School Boston Massachusetts USA
| |
Collapse
|
75
|
Zhang Y, Hong Z, Yuan Z, Wang T, Wu X, Liu B, Ai Z, Wu H, Yang Y. Extract from Rostellularia procumbens (L.) Nees Inhibits Thrombosis and Platelet Aggregation by Regulating Integrin β 3 and MAPK Pathways. ACS OMEGA 2020; 5:32123-32130. [PMID: 33344867 PMCID: PMC7745434 DOI: 10.1021/acsomega.0c05227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 11/23/2020] [Indexed: 06/12/2023]
Abstract
AIM OF STUDY The main objective of this study was to investigate the antithrombotic and antiplatelet effect of the extract from Rostellularia procumbenss (L.) Nees and understand the mechanisms by which it exerts its antithrombotic and antiplatelet mechanisms. MATERIALS AND METHODS The antithrombotic effective parts (RPE) were isolated using D101 macroporous adsorption resin and potential active ingredients (JAC) were isolated using the preparative liquid-phase method. The lactate dehydrogenase kit was used to determine the toxicity of RPE and JAC to platelets. The antiadhesion effect of RPE and JAC on platelets was observed by fluorescence microscopy with rhodamine phalloidin. Antithrombotic efficacy of RPE and JAC in vivo was evaluated by establishing a rat tail thrombosis model. Contents of p-selectin, TXB2, and 6-keto-PGF1α in rat serum were measured using an enzyme-linked immunosorbent (ELISA) assay, and the rat black tail rate was measured to prove the protective effect of RPE and JAC on the tail thrombus rat model. Western blot was used for detection of serum-related proteins in the tail thrombus rat model. RESULTS The results showed that RPE had antithrombotic and antiplatelet effects. RPE and JAC have no toxicity to platelets. In vitro experiments showed that RPE and JAC had antiadhesion effects on platelets. In vivo experiments showed that RPE significantly inhibited the increase of p-selectin and TXB2 and significantly increased the content of 6-keto-PGF1α in the serum of rats. Western blot results demonstrated that RPE and JDB significantly inhibited the phosphorylation of the MAPK protein family in the platelets of rats, and RPE also significantly inhibited the phosphorylation of β3 protein. CONCLUSIONS RPE has antithrombotic and antiplatelet activity in vivo and vitro. Its mechanism may be via preventing integrin αIIbβ3 activation, which in turn leads to the inhibition of the phosphorylation of the MAPK family and further suppresses TXA2, which leads to the antithrombotic and antiplatelet effects.
Collapse
Affiliation(s)
- Ying Zhang
- Faculty of Pharmacy, Hubei University of
Chinese Medicine, Wuhan 430065, China
| | - Zongchao Hong
- Faculty of Pharmacy, Hubei University of
Chinese Medicine, Wuhan 430065, China
| | - Zixin Yuan
- Faculty of Pharmacy, Hubei University of
Chinese Medicine, Wuhan 430065, China
| | - Tianshun Wang
- Faculty of Pharmacy, Hubei University of
Chinese Medicine, Wuhan 430065, China
| | - Xingpan Wu
- Faculty of Pharmacy, Hubei University of
Chinese Medicine, Wuhan 430065, China
| | - Bo Liu
- Faculty of Pharmacy, Hubei University of
Chinese Medicine, Wuhan 430065, China
| | - Zhongzhu Ai
- Faculty of Pharmacy, Hubei University of
Chinese Medicine, Wuhan 430065, China
| | - Hezhen Wu
- Faculty of Pharmacy, Hubei University of
Chinese Medicine, Wuhan 430065, China
- Key Laboratory
of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, Hubei University of Chinese Medicine, Wuhan 430065, China
- Collaborative Innovation Center of Traditional
Chinese Medicine of New Products for Geriatrics Hubei Province, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Yanfang Yang
- Faculty of Pharmacy, Hubei University of
Chinese Medicine, Wuhan 430065, China
- Key Laboratory
of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, Hubei University of Chinese Medicine, Wuhan 430065, China
- Collaborative Innovation Center of Traditional
Chinese Medicine of New Products for Geriatrics Hubei Province, Hubei University of Chinese Medicine, Wuhan 430065, China
| |
Collapse
|
76
|
Faria AVS, Andrade SS, Peppelenbosch MP, Ferreira-Halder CV, Fuhler GM. The role of phospho-tyrosine signaling in platelet biology and hemostasis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118927. [PMID: 33310067 DOI: 10.1016/j.bbamcr.2020.118927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 12/01/2020] [Accepted: 12/05/2020] [Indexed: 10/22/2022]
Abstract
Platelets are small enucleated cell fragments specialized in the control of hemostasis, but also playing a role in angiogenesis, inflammation and immunity. This plasticity demands a broad range of physiological processes. Platelet functions are mediated through a variety of receptors, the concerted action of which must be tightly regulated, in order to allow specific and timely responses to different stimuli. Protein phosphorylation is one of the main key regulatory mechanisms by which extracellular signals are conveyed. Despite the importance of platelets in health and disease, the molecular pathways underlying the activation of these cells are still under investigation. Here, we review current literature on signaling platelet biology and in particular emphasize the newly emerging role of phosphatases in these processes.
Collapse
Affiliation(s)
- Alessandra V S Faria
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands; Department of Biochemistry and Tissue Biology, University of Campinas, UNICAMP, Campinas, SP 13083-862, Brazil
| | | | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands
| | - Carmen V Ferreira-Halder
- Department of Biochemistry and Tissue Biology, University of Campinas, UNICAMP, Campinas, SP 13083-862, Brazil
| | - Gwenny M Fuhler
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands.
| |
Collapse
|
77
|
K. Poddar M, Banerjee S. Molecular Aspects of Pathophysiology of Platelet Receptors. Platelets 2020. [DOI: 10.5772/intechopen.92856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Receptor is a dynamic instrumental surface protein that helps to interact with specific molecules to respond accordingly. Platelet is the smallest in size among the blood components, but it plays many pivotal roles to maintain hemostasis involving its surface receptors. It (platelet) has cell adhesion receptors (e.g., integrins and glycoproteins), leucine-rich repeats receptors (e.g., TLRs, glycoprotein complex, and MMPs), selectins (e.g., CLEC, P-selectin, and CD), tetraspanins (e.g., CD and LAMP), transmembrane receptors (e.g., purinergic—P2Y and P2X1), prostaglandin receptors (e.g., TxA2, PGH2, and PGI2), immunoglobulin superfamily receptors (e.g., FcRγ and FcεR), etc. on its surface. The platelet receptors (e.g., glycoproteins, protease-activated receptors, and GPCRs) during platelet activation are over expressed and their granule contents are secreted (including neurotransmitters, cytokines, and chemokines) into circulation, which are found to be correlated with different physiological conditions. Interestingly, platelets promote metastasis through circulation protecting from cytolysis and endogenous immune surveillance involving several platelets receptors. The updated knowledge about different types of platelet receptors in all probable aspects, including their inter- and intra-signaling mechanisms, are discussed with respect to not only its (platelets) receptor type but also under different pathophysiological conditions.
Collapse
|
78
|
Mechanisms and biomarkers of cancer-associated thrombosis. Transl Res 2020; 225:33-53. [PMID: 32645431 PMCID: PMC8020882 DOI: 10.1016/j.trsl.2020.06.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/24/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023]
Abstract
Cancer-associated thrombosis is a leading cause of non-cancer death in cancer patients and is comprised of both arterial and venous thromboembolism (VTE). There are multiple risk factors for developing VTE, including cancer type, stage, treatment, and other medical comorbidities, which suggests that the etiology of thrombosis is multifactorial. While cancer-associated thrombosis can be treated with anticoagulation, benefits of therapy must be balanced with the increased bleeding risks seen in patients with cancer. Although risk models exist for primary and recurrent VTE, additional predictors are needed to improve model performance and discrimination of high-risk patients. This review will outline the diverse mechanisms driving thrombosis in cancer patients, as well as provide an overview of biomarkers studied in thrombosis risk and important considerations when selecting candidate biomarkers.
Collapse
|
79
|
Apte G, Börke J, Rothe H, Liefeith K, Nguyen TH. Modulation of Platelet-Surface Activation: Current State and Future Perspectives. ACS APPLIED BIO MATERIALS 2020; 3:5574-5589. [PMID: 35021790 DOI: 10.1021/acsabm.0c00822] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Modulation of platelet-surface activation is important for many biomedical applications such as in vivo performance, platelet storage, and acceptance of an implant. Reducing platelet-surface activation is challenging because they become activated immediately after short contact with nonphysiological surfaces. To date, controversies and open questions in the field of platelet-surface activation still remain. Here, we review state-of-the-art approaches in inhibiting platelet-surface activation, mainly focusing on modification, patterning, and methodologies for characterization of the surfaces. As a future perspective, we discuss how the combination of biochemical and physiochemical strategies together with the topographical modulations would assist in the search for an ideal nonthrombogenic surface.
Collapse
|
80
|
Yang Y, Wang B, Tian Q, Li B. Purification and Characterization of Novel Collagen Peptides against Platelet Aggregation and Thrombosis from Salmo salar. ACS OMEGA 2020; 5:19995-20003. [PMID: 32832753 PMCID: PMC7439260 DOI: 10.1021/acsomega.0c01340] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/06/2020] [Indexed: 06/11/2023]
Abstract
Collagen is a rich source of bioactive peptides and is widely distributed in the skin and bone tissue. In this study, collagen from Salmo salar skin was hydrolyzed with Alcalase or Protamex followed by simulated digestion, YMC ODS-A C18 separation, and ESI-MS/MS analysis. A total of 19 peptides were identified and synthesized for investigation of their antiplatelet activities. Hyp-Gly-Glu-Phe-Gly (OGEFG) and Asp-Glu-Gly-Pro (DEGP) exhibited the most potent activity against ADP-induced platelet aggregation among them with IC50 values of 277.17 and 290.00 μM, respectively, and inhibited the release of β-TG and 5-HT in a dose-dependent manner significantly. Single oral administration of OGEFG and DEGP also inhibited thrombus formation in a ferric chloride-induced arterial thrombosis model at a dose of 200 μmol/kg body weight and did not prolong the bleeding time or cause an immune response in mice. Therefore, our findings indicated that collagen peptides had a potential to be developed into an effective specific medical food in the prevention of thrombotic diseases.
Collapse
Affiliation(s)
- Yijie Yang
- College
of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Bo Wang
- College
of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Qi Tian
- College
of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Bo Li
- College
of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Key
Laboratory of Functional Dairy, Ministry of Education, Beijing 100083, China
| |
Collapse
|
81
|
Song W, Gottschalk CJ, Tang TX, Biscardi A, Ellena JF, Finkielstein CV, Brown AM, Capelluto DGS. Structural, in silico, and functional analysis of a Disabled-2-derived peptide for recognition of sulfatides. Sci Rep 2020; 10:13520. [PMID: 32782308 PMCID: PMC7421900 DOI: 10.1038/s41598-020-70478-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/16/2020] [Indexed: 01/08/2023] Open
Abstract
Disabled-2 (Dab2) is an adaptor protein that regulates the extent of platelet aggregation by two mechanisms. In the first mechanism, Dab2 intracellularly downregulates the integrin αIIbβ3 receptor, converting it to a low affinity state for adhesion and aggregation processes. In the second mechanism, Dab2 is released extracellularly and interacts with the pro-aggregatory mediators, the integrin αIIbβ3 receptor and sulfatides, blocking their association to fibrinogen and P-selectin, respectively. Our previous research indicated that a 35-amino acid region within Dab2, which we refer to as the sulfatide-binding peptide (SBP), contains two potential sulfatide-binding motifs represented by two consecutive polybasic regions. Using molecular docking, nuclear magnetic resonance, lipid-binding assays, and surface plasmon resonance, this work identifies the critical Dab2 residues within SBP that are responsible for sulfatide binding. Molecular docking suggested that a hydrophilic region, primarily mediated by R42, is responsible for interaction with the sulfatide headgroup, whereas the C-terminal polybasic region contributes to interactions with acyl chains. Furthermore, we demonstrated that, in Dab2 SBP, R42 significantly contributes to the inhibition of platelet P-selectin surface expression. The Dab2 SBP residues that interact with sulfatides resemble those described for sphingolipid-binding in other proteins, suggesting that sulfatide-binding proteins share common binding mechanisms.
Collapse
Affiliation(s)
- Wei Song
- Protein Signaling Domains Laboratory, Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Carter J Gottschalk
- Research and Informatics, University Libraries, Biochemistry Department, and Center for Drug Discovery, Virginia Tech, Blacksburg, 24061, VA, USA
| | - Tuo-Xian Tang
- Protein Signaling Domains Laboratory, Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Andrew Biscardi
- Protein Signaling Domains Laboratory, Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Jeffrey F Ellena
- Biomolecular Magnetic Resonance Facility, University of Virginia, Charlottesville, VA, 22904, USA
| | - Carla V Finkielstein
- Integrated Cellular Responses Laboratory, Fralin Biomedical Research Institute, Department of Biological Sciences, and Center for Drug Discovery, Virginia Tech, Roanoke, VA, 24016, USA
| | - Anne M Brown
- Research and Informatics, University Libraries, Biochemistry Department, and Center for Drug Discovery, Virginia Tech, Blacksburg, 24061, VA, USA
| | - Daniel G S Capelluto
- Protein Signaling Domains Laboratory, Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
82
|
Arenas Gómez CM, Sabin KZ, Echeverri K. Wound healing across the animal kingdom: Crosstalk between the immune system and the extracellular matrix. Dev Dyn 2020; 249:834-846. [PMID: 32314465 PMCID: PMC7383677 DOI: 10.1002/dvdy.178] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/10/2020] [Accepted: 04/11/2020] [Indexed: 12/11/2022] Open
Abstract
Tissue regeneration is widespread in the animal kingdom. To date, key roles for different molecular and cellular programs in regeneration have been described, but the ultimate blueprint for this talent remains elusive. In animals capable of tissue regeneration, one of the most crucial stages is wound healing, whose main goal is to close the wound and prevent infection. In this stage, it is necessary to avoid scar formation to facilitate the activation of the immune system and remodeling of the extracellular matrix, key factors in promoting tissue regeneration. In this review, we will discuss the current state of knowledge regarding the role of the immune system and the interplay with the extracellular matrix to trigger a regenerative response.
Collapse
Affiliation(s)
- Claudia M. Arenas Gómez
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological LaboratoryWoods HoleMassachusettsUSA
| | - Keith Z. Sabin
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological LaboratoryWoods HoleMassachusettsUSA
| | - Karen Echeverri
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological LaboratoryWoods HoleMassachusettsUSA
| |
Collapse
|
83
|
Palma-Barqueros V, Bohdan N, Revilla N, Vicente V, Bastida JM, Rivera J. PTGS1 gene variations associated with bleeding and platelet dysfunction. Platelets 2020; 32:710-716. [PMID: 32584621 DOI: 10.1080/09537104.2020.1782370] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | - Natalia Bohdan
- Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, Murcia, Spain
| | - Nuria Revilla
- Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, Murcia, Spain
| | - Vicente Vicente
- Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, Murcia, Spain
| | - José M Bastida
- Department of Hematology, IBSAL-Hospital Universitario de Salamanca, Salamanca, Spain.,On Behalf of the "Inherited Platelet Disorders Project", Hemorrhagic Diathesis Working Group, SETH
| | - José Rivera
- Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, Murcia, Spain.,On Behalf of the "Inherited Platelet Disorders Project", Hemorrhagic Diathesis Working Group, SETH
| |
Collapse
|
84
|
Reddoch-Cardenas KM, Sharma U, Salgado CL, Cantu C, Darlington DN, Pidcoke HF, Bynum JA, Cap AP. Use of Specialized Pro-Resolving Mediators to Alleviate Cold Platelet Storage Lesion. Transfusion 2020; 60 Suppl 3:S112-S118. [PMID: 32478925 DOI: 10.1111/trf.15750] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/19/2020] [Accepted: 02/24/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Cold-stored platelets are an attractive option for treatment of actively bleeding patients due to a reduced risk of septic complications and preserved hemostatic function compared to conventional room temperature-stored platelets. However, refrigeration causes increased platelet activation and aggregate formation. Specialized pro-resolving mediators (SPMs), cell signaling mediators biosynthesized from essential fatty acids, have been shown to modulate platelet function and activation. In this study, we sought to determine if SPMs could be used to inhibit cold-stored platelet activation. METHODS Platelets were collected from healthy donors (n = 4-7) and treated with SPMs (resolvin E1 [RvE1], maresin 1 [MaR1], and resolvin D2 [RvD2]) or vehicle (VEH; 0.1% EtOH). Platelets were stored without agitation in the cold and assayed on Days 0 and 7 of storage for platelet activation levels using flow cytometry, platelet count, aggregation response using impedance aggregometry, and nucleotide content using mass spectrometry. RESULTS Compared to VEH, SPM treatment inhibited GPIb shedding (all compounds), significantly reduced both PS exposure and activation of GPIIb/IIIa receptor (RvD2, MaR1), and preserved aggregation response to TRAP (RvD2, MaR1) after 7 days of storage. Similar to untreated cold-stored platelets, SPM-treated samples did not preserve platelet counts or block the release of P-Selectin. Nucleotide content was unaffected by SPM treatment in cold-stored platelets. CONCLUSIONS SPM treatment, particularly Mar1 and RvD2, led to reduced platelet activation and preserved platelet function after 7 days of storage in the cold. Future work is warranted to better elucidate the mechanism of action of SPMs on cold platelet function and activation.
Collapse
Affiliation(s)
- Kristin M Reddoch-Cardenas
- Coagulation and Blood Research Program, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Umang Sharma
- Coagulation and Blood Research Program, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Christi L Salgado
- Coagulation and Blood Research Program, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Carolina Cantu
- Coagulation and Blood Research Program, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Daniel N Darlington
- Coagulation and Blood Research Program, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Heather F Pidcoke
- Translational Medicine Institute, Colorado State University, Fort Collins, Colorado, USA
| | - James A Bynum
- Coagulation and Blood Research Program, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Andrew P Cap
- Coagulation and Blood Research Program, U.S. Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| |
Collapse
|
85
|
Zhou Y, Yasumoto A, Lei C, Huang CJ, Kobayashi H, Wu Y, Yan S, Sun CW, Yatomi Y, Goda K. Intelligent classification of platelet aggregates by agonist type. eLife 2020; 9:52938. [PMID: 32393438 PMCID: PMC7217700 DOI: 10.7554/elife.52938] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 04/15/2020] [Indexed: 12/18/2022] Open
Abstract
Platelets are anucleate cells in blood whose principal function is to stop bleeding by forming aggregates for hemostatic reactions. In addition to their participation in physiological hemostasis, platelet aggregates are also involved in pathological thrombosis and play an important role in inflammation, atherosclerosis, and cancer metastasis. The aggregation of platelets is elicited by various agonists, but these platelet aggregates have long been considered indistinguishable and impossible to classify. Here we present an intelligent method for classifying them by agonist type. It is based on a convolutional neural network trained by high-throughput imaging flow cytometry of blood cells to identify and differentiate subtle yet appreciable morphological features of platelet aggregates activated by different types of agonists. The method is a powerful tool for studying the underlying mechanism of platelet aggregation and is expected to open a window on an entirely new class of clinical diagnostics, pharmacometrics, and therapeutics. Platelets are small cells in the blood that primarily help stop bleeding after an injury by sticking together with other blood cells to form a clot that seals the broken blood vessel. Blood clots, however, can sometimes cause harm. For example, if a clot blocks the blood flow to the heart or the brain, it can result in a heart attack or stroke, respectively. Blood clots have also been linked to harmful inflammation and the spread of cancer, and there are now preliminary reports of remarkably high rates of clotting in COVID-19 patients in intensive care units. A variety of chemicals can cause platelets to stick together. It has long been assumed that it would be impossible to tell apart the clots formed by different chemicals (which are also known as agonists). This is largely because these aggregates all look very similar under a microscope, making it incredibly time consuming for someone to look at enough microscopy images to reliably identify the subtle differences between them. However, finding a way to distinguish the different types of platelet aggregates could lead to better ways to diagnose or treat blood vessel-clogging diseases. To make this possible, Zhou, Yasumoto et al. have developed a method called the “intelligent platelet aggregate classifier” or iPAC for short. First, numerous clot-causing chemicals were added to separate samples of platelets taken from healthy human blood. The method then involved using high-throughput techniques to take thousands of images of these samples. Then, a sophisticated computer algorithm called a deep learning model analyzed the resulting image dataset and “learned” to distinguish the chemical causes of the platelet aggregates based on subtle differences in their shapes. Finally, Zhou, Yasumoto et al. verified iPAC method’s accuracy using a new set of human platelet samples. The iPAC method may help scientists studying the steps that lead to clot formation. It may also help clinicians distinguish which clot-causing chemical led to a patient’s heart attack or stroke. This could help them choose whether aspirin or another anti-platelet drug would be the best treatment. But first more studies are needed to confirm whether this method is a useful tool for drug selection or diagnosis.
Collapse
Affiliation(s)
- Yuqi Zhou
- Department of Chemistry, University of Tokyo, Tokyo, Japan
| | - Atsushi Yasumoto
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Cheng Lei
- Department of Chemistry, University of Tokyo, Tokyo, Japan.,Institute of Technological Sciences, Wuhan University, Hubei, China
| | - Chun-Jung Huang
- Department of Photonics, National Chiao Tung University, Hsinchu, Taiwan
| | | | - Yunzhao Wu
- Department of Chemistry, University of Tokyo, Tokyo, Japan
| | - Sheng Yan
- Department of Chemistry, University of Tokyo, Tokyo, Japan
| | - Chia-Wei Sun
- Department of Photonics, National Chiao Tung University, Hsinchu, Taiwan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Keisuke Goda
- Department of Chemistry, University of Tokyo, Tokyo, Japan.,Institute of Technological Sciences, Wuhan University, Hubei, China.,Department of Bioengineering, University of California, Los Angeles, United States
| |
Collapse
|
86
|
Wei G, Xu X, Tong H, Wang X, Chen Y, Ding Y, Zhang S, Ju W, Fu C, Li Z, Zeng L, Xu K, Qiao J. Salidroside inhibits platelet function and thrombus formation through AKT/GSK3β signaling pathway. Aging (Albany NY) 2020; 12:8151-8166. [PMID: 32352928 PMCID: PMC7244060 DOI: 10.18632/aging.103131] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/30/2020] [Indexed: 12/17/2022]
Abstract
Salidroside is the main bioactive component in Rhodiola rosea and possesses multiple biological and pharmacological properties. However, whether salidroside affects platelet function remains unclear. Our study aims to investigate salidroside’s effect on platelet function. Human or mouse platelets were treated with salidroside (0-20 μM) for 1 hour at 37°C. Platelet aggregation, granule secretion, and receptors expression were measured together with detection of platelet spreading and clot retraction. In addition, salidroside (20 mg/kg) was intraperitoneally injected into mice followed by measuring tail bleeding time, arterial and venous thrombosis. Salidroside inhibited thrombin- or CRP-induced platelet aggregation and ATP release and did not affect the expression of P-selectin, glycoprotein (GP) Ibα, GPVI and αIIbβ3. Salidroside-treated platelets presented decreased spreading on fibrinogen or collagen and reduced clot retraction with decreased phosphorylation of c-Src, Syk and PLCγ2. Additionally, salidroside significantly impaired hemostasis, arterial and venous thrombus formation in mice. Moreover, in thrombin-stimulated platelets, salidroside inhibited phosphorylation of AKT (T308/S473) and GSK3β (Ser9). Further, addition of GSK3β inhibitor reversed the inhibitory effect of salidroside on platelet aggregation and clot retraction. In conclusion, salidroside inhibits platelet function and thrombosis via AKT/GSK3β signaling, suggesting that salidroside may be a novel therapeutic drug for treating thrombotic or cardiovascular diseases.
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, Xuzhou, China
| | - Xiaoqi 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, Xuzhou, China
| | - Huan Tong
- 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, 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, Xuzhou, China
| | - Yuting Chen
- 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, Xuzhou, China
| | - Yangyang Ding
- 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, Xuzhou, China
| | - Sixuan Zhang
- 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, 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, Xuzhou, China
| | - Chunling Fu
- 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, 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, Xuzhou, China
| | - Lingyu Zeng
- 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, 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, 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, Xuzhou, China
| |
Collapse
|
87
|
Saito H, Hayakawa M, Kamoshita N, Yasumoto A, Suzuki-Inoue K, Yatomi Y, Ohmori T. Establishment of a megakaryoblastic cell line for conventional assessment of platelet calcium signaling. Int J Hematol 2020; 111:786-794. [PMID: 32180119 DOI: 10.1007/s12185-020-02853-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/28/2020] [Accepted: 03/01/2020] [Indexed: 10/24/2022]
Abstract
Platelet function tests utilizing agonists or patient serum are generally performed to assess platelet activation ex vivo. However, inter-individual differences in platelet reactivity and donor requirements make it difficult to standardize these tests. Here, we established a megakaryoblastic cell line for the conventional assessment of platelet activation. We first compared intracellular signaling pathways using CD32 crosslinking in several megakaryoblastic cell lines, including CMK, UT-7/TPO, and MEG-01 cells. We confirmed that CD32 was abundantly expressed on the cell surface, and that intracellular calcium mobilization and tyrosine phosphorylation occurred after CD32 crosslinking. We next employed GCaMP6s, a highly sensitive calcium indicator, to facilitate the detection of calcium mobilization by transducing CMK and MEG-01 cells with a plasmid harboring GCaMP6s under the control of the human elongation factor-1α promoter. Cells that stably expressed GCaMP6s emitted enhanced green fluorescent protein fluorescence in response to intracellular calcium mobilization following agonist stimulation in the absence of pretreatment. In summary, we have established megakaryoblastic cell lines that mimic platelets by mobilizing intracellular calcium in response to several agonists. These cell lines can potentially be utilized in high-throughput screening assays for the discovery of new antiplatelet drugs or diagnosis of disorders caused by platelet-activating substances.
Collapse
Affiliation(s)
- Hiroshi Saito
- Department of Biochemistry, Jichi Medical University School of Medicine, Tochigi, 329-0498, Japan
| | - Morisada Hayakawa
- Department of Biochemistry, Jichi Medical University School of Medicine, Tochigi, 329-0498, Japan
| | - Nobuhiko Kamoshita
- Department of Biochemistry, Jichi Medical University School of Medicine, Tochigi, 329-0498, Japan
| | - Atsushi Yasumoto
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Katsue Suzuki-Inoue
- Department of Clinical and Laboratory Medicine, Faculty of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Tsukasa Ohmori
- Department of Biochemistry, Jichi Medical University School of Medicine, Tochigi, 329-0498, Japan.
| |
Collapse
|
88
|
Barale C, Russo I. Influence of Cardiometabolic Risk Factors on Platelet Function. Int J Mol Sci 2020; 21:ijms21020623. [PMID: 31963572 PMCID: PMC7014042 DOI: 10.3390/ijms21020623] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/16/2022] Open
Abstract
Platelets are key players in the thrombotic processes. The alterations of platelet function due to the occurrence of metabolic disorders contribute to an increased trend to thrombus formation and arterial occlusion, thus playing a major role in the increased risk of atherothrombotic events in patients with cardiometabolic risk factors. Several lines of evidence strongly correlate metabolic disorders such as obesity, a classical condition of insulin resistance, dyslipidemia, and impaired glucose homeostasis with cardiovascular diseases. The presence of these clinical features together with hypertension and disturbed microhemorrheology are responsible for the prothrombotic tendency due, at least partially, to platelet hyperaggregability and hyperactivation. A number of clinical platelet markers are elevated in obese and type 2 diabetes (T2DM) patients, including the mean platelet volume, circulating levels of platelet microparticles, oxidation products, platelet-derived soluble P-selectin and CD40L, thus contributing to an intersection between obesity, inflammation, and thrombosis. In subjects with insulin resistance and T2DM some defects depend on a reduced sensitivity to mediators—such as nitric oxide and prostacyclin—playing a physiological role in the control of platelet aggregability. Furthermore, other alterations occur only in relation to hyperglycemia. In this review, the main cardiometabolic risk factors, all components of metabolic syndrome involved in the prothrombotic tendency, will be taken into account considering some of the mechanisms involved in the alterations of platelet function resulting in platelet hyperactivation.
Collapse
|
89
|
Abstract
Von Willebrand factor (VWF) and coagulation factor VIII (FVIII) circulate as a complex in plasma and have a major role in the hemostatic system. VWF has a dual role in hemostasis. It promotes platelet adhesion by anchoring the platelets to the subendothelial matrix of damaged vessels and it protects FVIII from proteolytic degradation. Moreover, VWF is an acute phase protein that has multiple roles in vascular inflammation and is massively secreted from Weibel-Palade bodies upon endothelial cell activation. Activated FVIII on the other hand, together with coagulation factor IX forms the tenase complex, an essential feature of the propagation phase of coagulation on the surface of activated platelets. VWF deficiency, either quantitative or qualitative, results in von Willebrand disease (VWD), the most common bleeding disorder. The deficiency of FVIII is responsible for Hemophilia A, an X-linked bleeding disorder. Here, we provide an overview on the role of the VWF-FVIII interaction in vascular physiology.
Collapse
Affiliation(s)
- Klytaimnistra Kiouptsi
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstrasse 1, Building 708, 55131, Mainz, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstrasse 1, Building 708, 55131, Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Mainz, Germany.
| |
Collapse
|
90
|
Ed Nignpense B, Chinkwo KA, Blanchard CL, Santhakumar AB. Polyphenols: Modulators of Platelet Function and Platelet Microparticle Generation? Int J Mol Sci 2019; 21:ijms21010146. [PMID: 31878290 PMCID: PMC6981839 DOI: 10.3390/ijms21010146] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/19/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022] Open
Abstract
Platelets and platelet microparticles (PMPs) play a key role in the pathophysiology of vascular disorders such as coronary artery disease and stroke. In atherosclerosis, for example, the disruption of the plaque exposes endogenous agonists such as collagen, which activates platelets. Platelet hyper-activation and the high levels of PMPs generated in such situations pose a thrombotic risk that can lead to strokes or myocardial infarctions. Interestingly, dietary polyphenols are gaining much attention due to their potential to mimic the antiplatelet activity of treatment drugs such as aspirin and clopidogrel that target the glycoprotein VI (GPVI)-collagen and cyclooxygenease-1 (COX-1)-thromboxane platelet activation pathways respectively. Platelet function tests such as aggregometry and flow cytometry used to monitor the efficacy of antiplatelet drugs can also be used to assess the antiplatelet potential of dietary polyphenols. Despite the low bioavailability of polyphenols, several in vitro and dietary intervention studies have reported antiplatelet effects of polyphenols. This review presents a summary of platelet function in terms of aggregation, secretion, activation marker expression, and PMP release. Furthermore, the review will critically evaluate studies demonstrating the impact of polyphenols on aggregation and PMP release.
Collapse
Affiliation(s)
- Borkwei Ed Nignpense
- School of Biomedical Sciences, Charles Sturt University, Locked Bag 588, Wagga Wagga, NSW 2678, Australia; (B.E.N.); (K.A.C.); (C.L.B.)
| | - Kenneth A. Chinkwo
- School of Biomedical Sciences, Charles Sturt University, Locked Bag 588, Wagga Wagga, NSW 2678, Australia; (B.E.N.); (K.A.C.); (C.L.B.)
- Australian Research Council (ARC), Industrial Transformation Training Centre (ITTC) for Functional Grains, Graham Centre for Agricultural Innovation, Charles Sturt University, Wagga Wagga, NSW 2650, Australia
| | - Christopher L. Blanchard
- School of Biomedical Sciences, Charles Sturt University, Locked Bag 588, Wagga Wagga, NSW 2678, Australia; (B.E.N.); (K.A.C.); (C.L.B.)
- Australian Research Council (ARC), Industrial Transformation Training Centre (ITTC) for Functional Grains, Graham Centre for Agricultural Innovation, Charles Sturt University, Wagga Wagga, NSW 2650, Australia
| | - Abishek B. Santhakumar
- School of Biomedical Sciences, Charles Sturt University, Locked Bag 588, Wagga Wagga, NSW 2678, Australia; (B.E.N.); (K.A.C.); (C.L.B.)
- Australian Research Council (ARC), Industrial Transformation Training Centre (ITTC) for Functional Grains, Graham Centre for Agricultural Innovation, Charles Sturt University, Wagga Wagga, NSW 2650, Australia
- Correspondence: ; Tel.: +61-2-6933-2678
| |
Collapse
|
91
|
Ye T, Shi H, Phan-Thien N, Lim CT. The key events of thrombus formation: platelet adhesion and aggregation. Biomech Model Mechanobiol 2019; 19:943-955. [PMID: 31754949 DOI: 10.1007/s10237-019-01262-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 11/11/2019] [Indexed: 01/20/2023]
Abstract
Thrombus formation is a complex, dynamic and multistep process, involving biochemical reactions, mechanical stimulation, hemodynamics, and so on. In this study, we concentrate on its two crucial steps: (i) platelets adhered to a vessel wall, or simply platelet adhesion, and (ii) platelets clumping and arrested to the adherent platelets, named platelet aggregation. We report the first direct simulation of three modes of platelet adhesion, detachment, rolling adhesion and firm adhesion, as well as the formation, disintegration, arrestment and consolidation of platelet plugs. The results show that the bond dissociation in the detachment mode is mainly attributed to a high probability of rupturing bonds, such that any existing bond can be quickly ruptured and all bonds would be completely broken. In the rolling adhesion, however, it is mainly attributed to the strong traction from the shear flow or erythrocytes, causing that the bonds are ruptured at the trailing edge of the platelet. The erythrocytes play an important role in platelet activities, such as the formation, disintegration, arrestment and consolidation of platelet plugs. They exert an aggregate force on platelets, a repulsion at a near distance but an attraction at a far distance to the platelets. This aggregate force can promote platelets to form a plug and/or bring along a part of a platelet plug causing its disintegration. It also greatly influences the arrestment and consolidation of platelet plugs, together with the adhesive force from the thrombus.
Collapse
Affiliation(s)
- Ting Ye
- School of Mathematics, Jilin University, Qianjin Ave. 2699, Changchun, 130012, China.
| | - Huixin Shi
- School of Mathematics, Jilin University, Qianjin Ave. 2699, Changchun, 130012, China
| | - Nhan Phan-Thien
- Department of Mechanical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Chwee Teck Lim
- Department of Mechanical Engineering, National University of Singapore, Singapore, 117576, Singapore
| |
Collapse
|
92
|
Yadav VK, Singh PK, Agarwal V, Singh SK. Crosstalk between Platelet and Bacteria: A Therapeutic Prospect. Curr Pharm Des 2019; 25:4041-4052. [PMID: 31553286 DOI: 10.2174/1381612825666190925163347] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/13/2019] [Indexed: 12/16/2022]
Abstract
Platelets are typically recognized for their roles in the maintenance of hemostasis and vascular wall repair to reduce blood loss. Beyond hemostasis, platelets also play a critical role in pathophysiological conditions like atherosclerosis, stroke, thrombosis, and infections. During infection, platelets interact directly and indirectly with bacteria through a wide range of cellular and molecular mechanisms. Platelet surface receptors such as GPIbα, FcγRIIA, GPIIbIIIa, and TLRs, etc. facilitate direct interaction with bacterial cells. Besides, the indirect interaction between platelet and bacteria involves host plasma proteins such as von Willebrand Factor (vWF), fibronectin, IgG, and fibrinogen. Bacterial cells induce platelet activation, aggregation, and thrombus formation in the microvasculature. The activated platelets induce the Neutrophil Extracellular Traps (NETs) formation, which further contribute to thrombosis. Thus, platelets are extensively anticipated as vital immune modulator cells during infection, which may further lead to cardiovascular complications. In this review, we cover the interaction mechanisms between platelets and bacteria that may lead to the development of thrombotic disorders. Platelet receptors and other host molecules involved in such interactions can be used to develop new therapeutic strategies to combat against infection-induced cardiovascular complications. In addition, we highlight other receptor and enzyme targets that may further reduce infection-induced platelet activation and various pathological conditions.
Collapse
Affiliation(s)
- Vivek K Yadav
- Department of Biotechnology Motilal Nehru National Institute of Technology Allahabad, Prayagraj, India
| | - Pradeep K Singh
- Department of Biotechnology Motilal Nehru National Institute of Technology Allahabad, Prayagraj, India
| | - Vishnu Agarwal
- Department of Biotechnology Motilal Nehru National Institute of Technology Allahabad, Prayagraj, India
| | - Sunil K Singh
- Department of Animal Sciences, Central University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
93
|
Affiliation(s)
- Thomas A. Blair
- Center for Platelet Research Studies, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Andrew L. Frelinger
- Center for Platelet Research Studies, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
94
|
Rana A, Westein E, Niego B, Hagemeyer CE. Shear-Dependent Platelet Aggregation: Mechanisms and Therapeutic Opportunities. Front Cardiovasc Med 2019; 6:141. [PMID: 31620451 PMCID: PMC6763557 DOI: 10.3389/fcvm.2019.00141] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 09/03/2019] [Indexed: 01/04/2023] Open
Abstract
Cardiovascular diseases (CVD) are the number one cause of morbidity and death worldwide. As estimated by the WHO, the global death rate from CVD is 31% wherein, a staggering 85% results from stroke and myocardial infarction. Platelets, one of the key components of thrombi, have been well-investigated over decades for their pivotal role in thrombus development in healthy as well as diseased blood vessels. In hemostasis, when a vascular injury occurs, circulating platelets are arrested at the site of damage, where they are activated and aggregate to form hemostatic thrombi, thus preventing further bleeding. However, in thrombosis, pathological activation of platelets occurs, leading to uncontrolled growth of a thrombus, which in turn can occlude the blood vessel or embolize, causing downstream ischemic events. The molecular processes causing pathological thrombus development are in large similar to the processes controlling physiological thrombus formation. The biggest challenge of anti-thrombotics and anti-platelet therapeutics has been to decouple the pathological platelet response from the physiological one. Currently, marketed anti-platelet drugs are associated with major bleeding complications for this exact reason; they are not effective in targeting pathological thrombi without interfering with normal hemostasis. Recent studies have emphasized the importance of shear forces generated from blood flow, that primarily drive platelet activation and aggregation in thrombosis. Local shear stresses in obstructed blood vessels can be higher by up to two orders of magnitude as compared to healthy vessels. Leveraging abnormal shear forces in the thrombus microenvironment may allow to differentiate between thrombosis and hemostasis and develop shear-selective anti-platelet therapies. In this review, we discuss the influence of shear forces on thrombosis and the underlying mechanisms of shear-induced platelet activation. Later, we summarize the therapeutic approaches to target shear-sensitive platelet activation and pathological thrombus growth, with a particular focus on the shear-sensitive protein von Willebrand Factor (VWF). Inhibition of shear-specific platelet aggregation and targeted drug delivery may prove to be much safer and efficacious approaches over current state-of-the-art antithrombotic drugs in the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Akshita Rana
- Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Erik Westein
- Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Be'eri Niego
- Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Christoph E Hagemeyer
- Nanobiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
95
|
Wang H, Ye Y, Wan W, Wang L, Li R, Li L, Yang L, Yang L, Gu Y, Dong L, Meng Z. Xinmailong Modulates Platelet Function and Inhibits Thrombus Formation via the Platelet αIIbβ3-Mediated Signaling Pathway. Front Pharmacol 2019; 10:923. [PMID: 31507419 PMCID: PMC6716460 DOI: 10.3389/fphar.2019.00923] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 07/22/2019] [Indexed: 01/19/2023] Open
Abstract
Background: Xinmailong (XML), a bioactive composite extracted from Periplaneta americana, has been widely used to treat cardiovascular diseases such as congestive heart failure. However, it is unclear whether XML has antiplatelet and antithrombotic effects. Methods: The effects of XML on agonist-induced platelet aggregation, adhesion and spreading, granule secretion, integrin α II bβ3 activation, and thrombus formation were evaluated. Phosphorylation of Syk, PLCγ2, Akt, GSK3β, and MAPK signaling molecules was also studied on agonist-induced platelets. In addition, the antithrombotic effects of XML were observed in vivo using an acute pulmonary thrombosis mouse model. Results: XML dose-dependently inhibited in vitro platelet aggregation and granule secretion induced by thrombin, collagen, and arachidonic acid (AA). XML also greatly reduced platelet adhesion and spreading on both collagen- and fibrinogen-coated surfaces. Biochemical analysis revealed that XML inhibited thrombin-, collagen-, and AA-induced phosphorylation of Syk, PLCγ2, Akt, GSK3β, and MAPK. Additionally, XML significantly inhibited in vivo thrombus formation in a collagen–epinephrine-induced acute pulmonary thrombosis mouse model. Conclusions and General Significance: Here, we provide the first report showing that XML inhibits platelet function and that it possesses antithrombotic activity. This suggests that XML could be a potential therapeutic candidate to prevent or treat platelet-related cardiovascular diseases.
Collapse
Affiliation(s)
- Huawei Wang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yujia Ye
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wen Wan
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Luqiao Wang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ruijie Li
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Longjun Li
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lihong Yang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lai Yang
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yajuan Gu
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ling Dong
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhaohui Meng
- Laboratory of Molecular Cardiology, Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
96
|
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: 5.0] [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
|
97
|
Cuenca-Zamora EJ, Ferrer-Marín F, Rivera J, Teruel-Montoya R. Tubulin in Platelets: When the Shape Matters. Int J Mol Sci 2019; 20:E3484. [PMID: 31315202 PMCID: PMC6678703 DOI: 10.3390/ijms20143484] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/15/2019] [Accepted: 06/17/2019] [Indexed: 12/12/2022] Open
Abstract
Platelets are anuclear cells with a short lifespan that play an essential role in many pathophysiological processes, including haemostasis, inflammation, infection, vascular integrity, and metastasis. Billions of platelets are produced daily from megakaryocytes (platelet precursors). Despite this high production, the number of circulating platelets is stable and, under resting conditions, they maintain their typical discoid shape thanks to cytoskeleton proteins. The activation of platelets is associated with dynamic and rapid changes in the cytoskeleton. Two cytoskeletal polymer systems exist in megakaryocytes and platelets: actin filaments and microtubules, based on actin, and α- and β-tubulin heterodimers, respectively. Herein, we will focus on platelet-specific tubulins and their alterations and role of the microtubules skeleton in platelet formation (thrombopoiesis). During this process, microtubules mediate elongation of the megakaryocyte extensions (proplatelet) and granule trafficking from megakaryocytes to nascent platelets. In platelets, microtubules form a subcortical ring, the so-called marginal band, which confers the typical platelet discoid shape and is also responsible for changes in platelet morphology upon activation. Molecular alterations in the gene encoding β1 tubulin and microtubules post-translational modifications may result in quantitative or qualitative changes in tubulin, leading to altered cytoskeleton reorganization that may induce changes in the platelet number (thrombocytopenia), morphology or function. Consequently, β1-tubulin modifications may participate in pathological and physiological processes, such as development.
Collapse
Affiliation(s)
- Ernesto José Cuenca-Zamora
- Servicio de Hematología y Oncología Médica, Centro Regional de Hemodonación, Hospital Universitario Morales Meseguer, IMIB-Arrixaca, Red CIBERER CB15/00055, 30003 Murcia, Spain
| | - Francisca Ferrer-Marín
- Servicio de Hematología y Oncología Médica, Centro Regional de Hemodonación, Hospital Universitario Morales Meseguer, IMIB-Arrixaca, Red CIBERER CB15/00055, 30003 Murcia, Spain.
- Grado de Medicina, Universidad Católica San Antonio (UCAM), Campus de los Jerónimos, 30107 Murcia, Spain.
| | - José Rivera
- Servicio de Hematología y Oncología Médica, Centro Regional de Hemodonación, Hospital Universitario Morales Meseguer, IMIB-Arrixaca, Red CIBERER CB15/00055, 30003 Murcia, Spain
| | - Raúl Teruel-Montoya
- Servicio de Hematología y Oncología Médica, Centro Regional de Hemodonación, Hospital Universitario Morales Meseguer, IMIB-Arrixaca, Red CIBERER CB15/00055, 30003 Murcia, Spain
| |
Collapse
|
98
|
Qasim H, Karim ZA, Hernandez KR, Lozano D, Khasawneh FT, Alshbool FZ. Arhgef1 Plays a Vital Role in Platelet Function and Thrombogenesis. J Am Heart Assoc 2019; 8:e011712. [PMID: 30994039 PMCID: PMC6512111 DOI: 10.1161/jaha.118.011712] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/25/2019] [Indexed: 01/09/2023]
Abstract
Background Platelets are the cellular mediators of hemostasis and thrombosis, and their function is regulated by a number of molecular mediators, such as small GTP ases. These small GTP ases are themselves regulated by guanine nucleotide exchange factors such as Arhgefs, several of which are found in platelets, including the highly expressed Arhgef1. However, the role of Arhgef1 in platelets has not yet been investigated. Methods and Results We employed mice with genetic deletion of Arhgef1 (ie, Arhgef1-/-) and investigated their platelet phenotype by employing a host of in vivo and in vitro platelet assays. Our results indicate that Arhgef1-/- mice had prolonged carotid artery occlusion and tail bleeding times. Moreover, platelets from these mice exhibited defective aggregation, dense and α granule secretion, α II bβ3 integrin activation, clot retraction and spreading, in comparison to their wild-type littermates. Finally, we also found that the mechanism by which Arhgef1 regulates platelets is mediated in part by a defect in the activation of the RhoA-Rho-associated kinase axis, but not Rap1b. Conclusions Our data demonstrate, for the first time, that Arhgef1 plays a critical role in platelet function, in vitro and in vivo.
Collapse
Affiliation(s)
- Hanan Qasim
- Department of Pharmaceutical SciencesSchool of PharmacyThe University of Texas El PasoEl PasoTX
| | - Zubair A. Karim
- Department of Pharmaceutical SciencesSchool of PharmacyThe University of Texas El PasoEl PasoTX
| | - Keziah R. Hernandez
- Department of Pharmaceutical SciencesSchool of PharmacyThe University of Texas El PasoEl PasoTX
| | | | - Fadi T. Khasawneh
- Department of Pharmaceutical SciencesSchool of PharmacyThe University of Texas El PasoEl PasoTX
| | - Fatima Z. Alshbool
- Department of Pharmaceutical SciencesSchool of PharmacyThe University of Texas El PasoEl PasoTX
| |
Collapse
|
99
|
Shu G, Chen Y, Liu T, Ren S, Kong Y. Antimicrobial Peptide Cathelicidin-BF Inhibits Platelet Aggregation by Blocking Protease-Activated Receptor 4. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-018-9677-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
100
|
Lu J, Hu P, Wei G, Luo Q, Qiao J, Geng D. Effect of alteplase on platelet function and receptor expression. J Int Med Res 2019; 47:1731-1739. [PMID: 30799665 PMCID: PMC6460619 DOI: 10.1177/0300060519829991] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Objective To investigate the role of alteplase, a widely-used thrombolytic drug, in platelet function. Methods Human platelets were incubated with different concentrations of alteplase followed by analysis of platelet aggregation in response to adenosine diphosphate (ADP), collagen, ristocetin, arachidonic acid or epinephrine using light transmittance aggregometry. Platelet activation and surface levels of platelet receptors GPIbα, GPVI and αIIbβ3 were analysed using flow cytometry. The effect of alteplase on clot retraction was also examined. Results This study demonstrated that alteplase significantly inhibited platelet aggregation in response to ADP, collagen and epinephrine in a dose-dependent manner, but it did not affect ristocetin- or arachidonic acid-induced platelet aggregation. Alteplase did not affect platelet activation as demonstrated by no differences in P-selectin levels and PAC-1 binding being observed in collagen-stimulated platelets after alteplase treatment compared with vehicle. There were no changes in the surface levels of the platelet receptors GPIbα, GPVI and αIIbβ3 in alteplase-treated platelets. Alteplase treatment reduced thrombin-mediated clot retraction. Conclusions Alteplase inhibits platelet aggregation and clot retraction without affecting platelet activation and surface receptor levels.
Collapse
Affiliation(s)
- Jun Lu
- 1 The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China.,2 Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China.,*These authors contributed equally to this study
| | - Peng Hu
- 2 Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China.,*These authors contributed equally to this study
| | - Guangyu Wei
- 3 Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Qi Luo
- 3 Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Jianlin Qiao
- 3 Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Deqin Geng
- 1 The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China.,2 Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| |
Collapse
|