1
|
Nash J, Pym D, Davies A, Saunders C, George C, Williams JO, Grinberg OY, James PE. Enhanced oxygen availability and preserved aggregative function in platelet concentrates stored at reduced platelet concentration. Transfusion 2025; 65:575-587. [PMID: 39673310 PMCID: PMC11925141 DOI: 10.1111/trf.18101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Storage of platelets as platelet concentrates for transfusion is limited to 7 days in the United Kingdom due to deleterious effects on platelet quality and function that occur over time. Oxygen (O2) availability and sufficient gaseous exchange are known to be essential in maintaining the viability and function of platelets stored for transfusion. Despite this, there is a paucity of studies undertaking direct measures of O2 and optimization of conditions throughout storage. We address this and modulate the storage conditions to improve platelet quality and function. STUDY DESIGN AND METHODS Electron paramagnetic resonance oximetry was implemented to directly measure the [O2] experienced by stored platelet concentrates and the O2 consumption rate under standard blood banking conditions. From these direct measures the mathematical modeling was then applied to predict the main parameters contributing to effective O2 distribution throughout the unit. RESULTS This study demonstrates reducing the storage [O2] to reflect near physiological levels significantly alters O2 distribution within the unit and negatively impacts platelet functionality and quality, and therefore is not a viable storage option. DISCUSSION We show the reduction of platelet concentration within a unit improves O2 availability and pH, promotes a more uniform distribution of O2 throughout prolonged storage, and maintains platelet agonist-induced aggregation comparable to 100% platelet concentration. This may be a viable option and could potentially lead to reduced donor demand.
Collapse
Affiliation(s)
- Jamie Nash
- Center for Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, UK
- Component Development and Research Laboratory, Welsh Blood Service NHS, Wales, UK
| | - Dean Pym
- Center for Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, UK
| | - A Davies
- Center for Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, UK
| | - Christine Saunders
- Component Development and Research Laboratory, Welsh Blood Service NHS, Wales, UK
| | - Chloe George
- Component Development and Research Laboratory, Welsh Blood Service NHS, Wales, UK
| | - J O Williams
- Center for Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, UK
| | - O Y Grinberg
- Dartmouth Medical School, Hanover, New Hampshire, USA
| | - Philip E James
- Center for Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, UK
| |
Collapse
|
2
|
Pym D, Davies AJ, Williams JO, Saunders C, George CE, James PE. Small volume platelet concentrates for neonatal use are more susceptible to shear-induced storage lesion. Platelets 2024; 35:2389967. [PMID: 39169763 DOI: 10.1080/09537104.2024.2389967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024]
Abstract
The impact of the biophysical environment on the platelet storage lesion (PSL) has mainly focused on reduced temperature storage, overlooking the significance of storage-induced shear stress. Shear stress in platelet storage refers to the frictional force acting parallel to the bag surface and exists solely through the implementation of agitation. This study investigates whether minimizing exposure to agitation-induced shear stress can alleviate the unexplained loss of function in stored platelet concentrates for neonatal transfusion (neonatal PCs). Using particle tracking analysis, fluid motion was measured in neonatal and adult platelet storage bags under agitation frequencies ranging from 20-60 rpm. Platelets stored at 20-60 rpm agitation over 8 days were examined by biochemical analysis, aggregation, and expression of activation markers. Results indicate that neonatal PCs experience significantly higher storage-induced shear stress compared to adult doses, leading to reduced functionality and increased activation from day 2 of storage. Adjusting the neonatal PC agitation frequency to 20 rpm improved functionality in early storage, while 40 rpm maintains this improvement throughout storage with reduced activation, compared to 60 rpm storage. This study confirms that small volume PC storage for neonatal use contributes to the PSL through the induction of shear stress, suggesting further evaluation of the recommended agitation frequency for neonatal PCs or postponement of the production of neonatal PCs until requested for neonatal transfusion.
Collapse
Affiliation(s)
- Dean Pym
- Centre of Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, Wales, UK
- Welsh Blood Service, Component Development and Research Laboratory, Pontyclun, Wales, UK
| | - Amanda J Davies
- Centre of Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, Wales, UK
| | - Jessica O Williams
- Centre of Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, Wales, UK
| | - Christine Saunders
- Welsh Blood Service, Component Development and Research Laboratory, Pontyclun, Wales, UK
| | - Chloë E George
- Welsh Blood Service, Component Development and Research Laboratory, Pontyclun, Wales, UK
| | - Philip E James
- Centre of Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, Wales, UK
| |
Collapse
|
3
|
Benson TW, Pike MM, Spuzzillo A, Hicks SM, Ali S, Pham M, Mix DS, Brunner SI, Wadding-Lee C, Conrad KA, Russell HM, Jennings C, Coughlin TM, Aggarwal A, Lyden S, Mani K, Björck M, Wanhainen A, Bhandari R, Lipworth-Elliot L, Robinson-Cohen C, Caputo FJ, Shim S, Quesada O, Tourdot B, Edwards TL, Tranter M, Gardiner EE, Mackman N, Cameron SJ, Owens AP. Soluble glycoprotein VI predicts abdominal aortic aneurysm growth rate and is a novel therapeutic target. Blood 2024; 144:1663-1678. [PMID: 38900973 PMCID: PMC11522893 DOI: 10.1182/blood.2023021655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 03/05/2024] [Accepted: 05/02/2024] [Indexed: 06/22/2024] Open
Abstract
ABSTRACT A common feature in patients with abdominal aortic aneurysms (AAAs) is the formation of a nonocclusive intraluminal thrombus (ILT) in regions of aortic dilation. Platelets are known to maintain hemostasis and propagate thrombosis through several redundant activation mechanisms, yet the role of platelet activation in the pathogenesis of AAA-associated ILT is still poorly understood. Thus, we sought to investigate how platelet activation affects the pathogenesis of AAA. Using RNA sequencing, we identified that the platelet-associated transcripts are significantly enriched in the ILT compared with the adjacent aneurysm wall and healthy control aortas. We found that the platelet-specific receptor glycoprotein VI (GPVI) is among the top enriched genes in AAA ILT and is increased on the platelet surface of patients with AAAs. Examination of a specific indicator of platelet activity, soluble GPVI (sGPVI), in 2 independent cohorts of patients with AAAs is highly predictive of an AAA diagnosis and associates more strongly with aneurysm growth rate than D-dimer in humans. Finally, intervention with the anti-GPVI antibody (JAQ1) in mice with established aneurysms blunted the progression of AAA in 2 independent mouse models. In conclusion, we show that the levels of sGPVI in humans can predict a diagnosis of AAA and AAA growth rate, which may be critical in the identification of high-risk patients. We also identify GPVI as a novel platelet-specific AAA therapeutic target, with minimal risk of adverse bleeding complications, for which none currently exists.
Collapse
Affiliation(s)
- Tyler W. Benson
- Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Mindy M. Pike
- Division of Epidemiology, Vanderbilt Genetics Institute, Institute of Medicine and Public Health, Nashville, TN
- Division of Nephrology and Hypertension, Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN
| | - Anthony Spuzzillo
- Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH
- Departments of Internal Medicine and Pathology and Laboratory Medicine, Pathobiology and Molecular Medicine Graduate Program, University of Cincinnati, Cincinnati, OH
| | - Sarah M. Hicks
- Division of Genome Science and Cancer, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Sidra Ali
- Division of Genome Science and Cancer, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Michael Pham
- Department of Cardiovascular Medicine, Section of Vascular Medicine, Heart and Vascular Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Doran S. Mix
- Division of Vascular Surgery, Department of Surgery, University of Rochester School of Medicine, Rochester, NY
| | - Seth I. Brunner
- Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Caris Wadding-Lee
- Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH
- Departments of Internal Medicine and Pathology and Laboratory Medicine, Pathobiology and Molecular Medicine Graduate Program, University of Cincinnati, Cincinnati, OH
| | - Kelsey A. Conrad
- Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH
- Departments of Internal Medicine and Pathology and Laboratory Medicine, Pathobiology and Molecular Medicine Graduate Program, University of Cincinnati, Cincinnati, OH
| | - Hannah M. Russell
- Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH
- Departments of Internal Medicine and Pathology and Laboratory Medicine, Pathobiology and Molecular Medicine Graduate Program, University of Cincinnati, Cincinnati, OH
| | - Courtney Jennings
- Department of Cardiovascular Medicine, Section of Vascular Medicine, Heart and Vascular Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Taylor M. Coughlin
- Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH
- Departments of Internal Medicine and Pathology and Laboratory Medicine, Pathobiology and Molecular Medicine Graduate Program, University of Cincinnati, Cincinnati, OH
| | - Anu Aggarwal
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Sean Lyden
- Department of Cardiovascular Medicine, Section of Vascular Medicine, Heart and Vascular Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Kevin Mani
- Department of Surgical Sciences, Vascular Surgery, Uppsala University, Uppsala, Sweden
| | - Martin Björck
- Department of Surgical Sciences, Vascular Surgery, Uppsala University, Uppsala, Sweden
| | - Anders Wanhainen
- Department of Surgical Sciences, Vascular Surgery, Uppsala University, Uppsala, Sweden
- Department of Surgical and Perioperative Sciences, Umeå University, Umeå, Sweden
| | - Rohan Bhandari
- Department of Cardiovascular Medicine, Section of Vascular Medicine, Heart and Vascular Institute, Cleveland Clinic Foundation, Cleveland, OH
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Loren Lipworth-Elliot
- Division of Epidemiology, Vanderbilt Genetics Institute, Institute of Medicine and Public Health, Nashville, TN
- Division of Nephrology and Hypertension, Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN
| | - Cassianne Robinson-Cohen
- Division of Nephrology and Hypertension, Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt-O'Brien Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN
| | - Francis J. Caputo
- Department of Cardiovascular Medicine, Section of Vascular Medicine, Heart and Vascular Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Sharon Shim
- Department of Cardiovascular Medicine, Section of Vascular Medicine, Heart and Vascular Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Odayme Quesada
- Women’s Heart Center, The Christ Hospital Heart and Vascular Institute, Cincinnati, OH
- The Carl and Edyth Lindner Center for Research and Education, The Christ Hospital, Cincinnati, OH
| | - Benjamin Tourdot
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Todd L. Edwards
- Division of Epidemiology, Vanderbilt Genetics Institute, Institute of Medicine and Public Health, Nashville, TN
| | - Michael Tranter
- Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Elizabeth E. Gardiner
- Division of Genome Science and Cancer, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Nigel Mackman
- Division of Hematology and Oncology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Scott J. Cameron
- Department of Cardiovascular Medicine, Section of Vascular Medicine, Heart and Vascular Institute, Cleveland Clinic Foundation, Cleveland, OH
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH
- Department of Hematology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - A. Phillip Owens
- Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
4
|
Vidallon MLP, Moon MJ, Liu H, Song Y, Crawford S, Teo BM, McFadyen JD, Bishop AI, Tabor RF, Peter K, Wang X. Engineering Hyperechogenic Colloids with Clot-Targeting Capabilities from Platelet-Derived Membranes. ACS APPLIED MATERIALS & INTERFACES 2024; 16. [PMID: 39360874 PMCID: PMC11492166 DOI: 10.1021/acsami.4c12024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Thrombosis-related cardiovascular diseases remain the leading global cause of mortality and morbidity. In this study, we present a pioneering approach in the field of nanobiotechnology, with a focus on clinical translation, aimed at advancing early diagnosis and enhancing treatment options for thrombotic disorders. We introduce the fabrication of Platelet Membrane-Derived Bubbles (PMBs), which exhibit distinctive characteristics compared to conventional nanoparticles. These PMBs possess an average diameter of 700 nm and a negative ζ-potential, mirroring the attributes of parent platelet membranes. Utilizing diagnostic ultrasound imaging, we demonstrated the ability to visualize PMBs as hyperechogenic entities in agarose phantoms in vitro and in live mice in vivo. Furthermore, through confocal laser microscopy, we verified the retention of crucial transmembrane proteins, such as CD41 (GPIIb) and CD42 (GPIb), pivotal in conferring platelet-specific targeting functions. Importantly, our platelet aggregation studies confirmed that PMBs do not induce platelet aggregation but instead adhere to preformed platelet-rich in vitro thrombi. Overall, our work showcases the safe and precise utilization of PMBs to directly target acute thrombosis induced by laser injury in murine mesenteric veins in vivo, as visualized through intravital microscopy. In conclusion, we have successfully developed a rapid method for generating PMBs with unique ultrasound-directed and thrombus-targeting properties. These exceptional attributes of PMBs hold significant promise for advancing the field of ultrasound diagnostic thrombus imaging and clot-targeted therapy in the clinical context.
Collapse
Affiliation(s)
- Mark Louis P. Vidallon
- Molecular
Imaging and Theranostics Laboratory, Baker
Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Parkville, VIC 3010, Australia
- School
of Chemistry, Monash University, Clayton, VIC 3800, Australia
- Baker
Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia
| | - Mitchell J. Moon
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Parkville, VIC 3010, Australia
- Atherothrombosis
and Vascular Biology Laboratory, Baker Heart
and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Haikun Liu
- Molecular
Imaging and Theranostics Laboratory, Baker
Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Parkville, VIC 3010, Australia
| | - Yuyang Song
- Molecular
Imaging and Theranostics Laboratory, Baker
Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Parkville, VIC 3010, Australia
| | - Simon Crawford
- Ramaciotti
Centre for Cryo-electron Microscopy, Monash
University, Clayton, VIC 3800, Australia
| | - Boon Mian Teo
- School
of Chemistry, Monash University, Clayton, VIC 3800, Australia
| | - James D. McFadyen
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Parkville, VIC 3010, Australia
- Atherothrombosis
and Vascular Biology Laboratory, Baker Heart
and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Department
of Clinical Hematology, The Alfred Hospital, Melbourne, VIC 3004, Australia
- School
of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Alexis I. Bishop
- School
of Physics and Astronomy, Monash University, Clayton, VIC 3800, Australia
| | - Rico F. Tabor
- School
of Chemistry, Monash University, Clayton, VIC 3800, Australia
| | - Karlheinz Peter
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Parkville, VIC 3010, Australia
- Baker
Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia
- Atherothrombosis
and Vascular Biology Laboratory, Baker Heart
and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- School
of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Xiaowei Wang
- Molecular
Imaging and Theranostics Laboratory, Baker
Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker
Department of Cardiometabolic Health, University
of Melbourne, Parkville, VIC 3010, Australia
- Baker
Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia
- School
of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| |
Collapse
|
5
|
Colon Hidalgo D, Jordan M, Posey JN, Burciaga SD, Nguyen TTN, Sul C, Lewis CV, Delaney C, Nozik ES. Lung EC-SOD Overexpression Prevents Hypoxia-Induced Platelet Activation and Lung Platelet Accumulation. Antioxidants (Basel) 2024; 13:975. [PMID: 39199221 PMCID: PMC11351248 DOI: 10.3390/antiox13080975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/01/2024] Open
Abstract
Pulmonary hypertension (PH) is a progressive disease marked by pulmonary vascular remodeling and right ventricular failure. Inflammation and oxidative stress are critical in PH pathogenesis, with early pulmonary vascular inflammation preceding vascular remodeling. Extracellular superoxide dismutase (EC-SOD), a key vascular antioxidant enzyme, mitigates oxidative stress and protects against inflammation and fibrosis in diverse lung and vascular disease models. This study utilizes a murine hypobaric hypoxia model to investigate the role of lung EC-SOD on hypoxia-induced platelet activation and platelet lung accumulation, a critical factor in PH-related inflammation. We found that lung EC-SOD overexpression blocked hypoxia-induced platelet activation and platelet accumulation in the lung. Though lung EC-SOD overexpression increased lung EC-SOD content, it did not impact plasma extracellular SOD activity. However, ex vivo, exogenous extracellular SOD treatment specifically blunted convulxin-induced platelet activation but did not blunt platelet activation with thrombin or ADP. Our data identify platelets as a novel target of EC-SOD in response to hypoxia, providing a foundation to advance the understanding of dysregulated redox signaling and platelet activation in PH and other chronic hypoxic lung diseases.
Collapse
Affiliation(s)
- Daniel Colon Hidalgo
- Department of Medicine, Division of Pulmonary and Critical Care, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Mariah Jordan
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Janelle N. Posey
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Samuel D. Burciaga
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Thi-Tina N. Nguyen
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Christina Sul
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Division of Critical Care, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Caitlin V. Lewis
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Division of Critical Care, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Cassidy Delaney
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Eva S. Nozik
- Cardiovascular Pulmonary Research Group, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Division of Critical Care, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
6
|
Jiang H, Nechipurenko DY, Panteleev MA, Xu K, Qiao J. Redox regulation of platelet function and thrombosis. J Thromb Haemost 2024; 22:1550-1557. [PMID: 38460839 DOI: 10.1016/j.jtha.2024.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/15/2024] [Accepted: 02/24/2024] [Indexed: 03/11/2024]
Abstract
Platelets are well-known players in several cardiovascular diseases such as atherosclerosis and venous thrombosis. There is increasing evidence demonstrating that reactive oxygen species (ROS) are generated within activated platelets. Nicotinamide adenine dinucleotide phosphate oxidase (NOX) is a major source of ROS generation in platelets. Ligand binding to platelet receptor glycoprotein (GP) VI stimulates intracellular ROS generation consisting of a spleen tyrosine kinase-independent production involving NOX activation and a following spleen tyrosine kinase-dependent generation. In addition to GPVI, stimulation of platelet thrombin receptors (protease-activated receptors [PARs]) can also trigger NOX-derived ROS production. Our recent study found that mitochondria-derived ROS production can be induced by engagement of thrombin receptors but not by GPVI, indicating that mitochondria are another source of PAR-dependent ROS generation apart from NOX. However, mitochondria are not involved in GPVI-dependent ROS generation. Once generated, the intracellular ROS are also involved in modulating platelet function and thrombus formation; therefore, the site-specific targeting of ROS production or clearance of excess ROS within platelets is a potential intervention and treatment option for thrombotic events. In this review, we will summarize the signaling pathways involving regulation of platelet ROS production and their role in platelet function and thrombosis, with a focus on GPVI- and PAR-dependent platelet responses.
Collapse
Affiliation(s)
- Huimin Jiang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Dmitry Yu Nechipurenko
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia; Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Science, Moscow, Russia; Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Mikhail A Panteleev
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia; Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Science, Moscow, Russia; Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.
| |
Collapse
|
7
|
Garzon Dasgupta AK, Martyanov AA, Ignatova AA, Zgoda VG, Novichkova GA, Panteleev MA, Sveshnikova AN. Comparison of platelet proteomic profiles between children and adults reveals origins of functional differences. Pediatr Res 2024; 95:966-973. [PMID: 37872237 DOI: 10.1038/s41390-023-02865-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/25/2023] [Accepted: 09/26/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND Platelets are blood cells responsible for the prevention of blood loss upon vessel wall disruption. It has been demonstrated that platelet functioning differs significantly between adult and pediatric donors. This study aimed to identify potential differences between the protein composition of platelets of pediatric, adolescent, and adult donors. METHODS Platelet functional testing was conducted with live cell flow cytometry. Using a straightforward approach to platelet washing based on the sequential platelets centrifugation-resuspension, we were able to obtain stable and robust proteomics results, which corresponded to previously published data. RESULTS We have identified that pediatric donors' platelets have increased amounts of proteins, responsible for mitochondrial activity, proteasome activity, and vesicle transport. Flow cytometry analysis of platelet intracellular signaling and functional responses revealed that platelets of the pediatric donors have diminished granule secretion and increased quiescent platelet calcium concentration and decreased calcium mobilization in response to ADP. We could explain the observed changes in calcium responses by the increased mitochondria protein content, and the changes in granule secretion could be explained by the differences in vesicle transport protein content. CONCLUSIONS Therefore, we can conclude that the age-dependence of platelet functional responses originates from the difference in platelet protein content. IMPACT Platelets of infants are known to functionally differ from the platelet of adult donors, although the longevity and persistivity of these differences are debatable. Pediatric donor platelets have enhanced amounts of mitochondrial, proteasomal, and vesicle transport proteins. Platelets of the pediatric donors had increased cytosolic calcium in the resting state, what is explained by the increased numbers of mitochondrial proteins. Infants had decreased platelet granule release, which resolved upon adolescence. Thus, platelets of the infants should be assessed differently from adult platelets. Differences in platelet proteomic contents persisted in adolescent groups, yet, no significant differences in platelet function were observed.
Collapse
Affiliation(s)
- Andrei K Garzon Dasgupta
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow, 109029, Russia
| | - Alexey A Martyanov
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow, 109029, Russia
- National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow, 117198, Russia
| | - Anastasia A Ignatova
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow, 109029, Russia
- National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow, 117198, Russia
| | - Victor G Zgoda
- Institute of Biomedical Chemistry, 10 bld. 8, Pogodinskaya str., 119121, Moscow, Russia
| | - Galina A Novichkova
- National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow, 117198, Russia
| | - Mikhail A Panteleev
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow, 109029, Russia
- National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow, 117198, Russia
- Lomonosov Moscow State University, 1/2 Leninskie gory, Moscow, 119991, Russia
| | - Anastasia N Sveshnikova
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow, 109029, Russia.
- National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow, 117198, Russia.
- Lomonosov Moscow State University, 1/2 Leninskie gory, Moscow, 119991, Russia.
| |
Collapse
|
8
|
Gilly A, Park YC, Tsafantakis E, Karaleftheri M, Dedoussis G, Zeggini E. Genome-wide meta-analysis of 92 cardiometabolic protein serum levels. Mol Metab 2023; 78:101810. [PMID: 37778719 PMCID: PMC10582065 DOI: 10.1016/j.molmet.2023.101810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/11/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023] Open
Abstract
OBJECTIVES Global cardiometabolic disease prevalence has grown rapidly over the years, making it the leading cause of death worldwide. Proteins are crucial components in biological pathways dysregulated in disease states. Identifying genetic components that influence circulating protein levels may lead to the discovery of biomarkers for early stages of disease or offer opportunities as therapeutic targets. METHODS Here, we carry out a genome-wide association study (GWAS) utilising whole genome sequencing data in 3,005 individuals from the HELIC founder populations cohort, across 92 proteins of cardiometabolic relevance. RESULTS We report 322 protein quantitative trait loci (pQTL) signals across 92 proteins, of which 76 are located in or near the coding gene (cis-pQTL). We link those association signals with changes in protein expression and cardiometabolic disease risk using colocalisation and Mendelian randomisation (MR) analyses. CONCLUSIONS The majority of previously unknown signals we describe point to proteins or protein interactions involved in inflammation and immune response, providing genetic evidence for the contributing role of inflammation in cardiometabolic disease processes.
Collapse
Affiliation(s)
- Arthur Gilly
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Young-Chan Park
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | | | | | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, Athens, Greece
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; Technical University of Munich (TUM) and Klinikum Rechts der Isar, TUM School of Medicine, Munich, Germany.
| |
Collapse
|
9
|
Rauch A, Dupont A, Rosa M, Desvages M, Le Tanno C, Abdoul J, Didelot M, Ung A, Ruez R, Jeanpierre E, Daniel M, Corseaux D, Spillemaeker H, Labreuche J, Pradines B, Rousse N, Lenting PJ, Moussa MD, Vincentelli A, Bordet JC, Staels B, Vincent F, Denis CV, Van Belle E, Casari C, Susen S. Shear Forces Induced Platelet Clearance Is a New Mechanism of Thrombocytopenia. Circ Res 2023; 133:826-841. [PMID: 37883587 DOI: 10.1161/circresaha.123.322752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Thrombocytopenia has been consistently described in patients with extracorporeal membrane oxygenation (ECMO) and associated with poor outcome. However, the prevalence and underlying mechanisms remain largely unknown, and a device-related role of ECMO in thrombocytopenia has been hypothesized. This study aims to investigate the mechanisms underlying thrombocytopenia in ECMO patients. METHODS In a prospective cohort of 107 ECMO patients, we investigated platelet count, functions, and glycoprotein shedding. In an ex vivo mock circulatory ECMO loop, we assessed platelet responses and VWF (von Willebrand factor)-GP Ibα (glycoprotein Ibα) interactions at low- and high-flow rates, in the presence or absence of red blood cells. The clearance of human platelets subjected or not to ex vivo perfusion was studied using an in vivo transfusion model in NOD/SCID (nonobese diabetic/severe combined Immunodeficient) mice. RESULTS In ECMO patients, we observed a time-dependent decrease in platelet count starting 1 hour after device onset, with a mean drop of 7%, 35%, and 41% at 1, 24, and 48 hours post-ECMO initiation (P=0.00013, P<0.0001, and P<0.0001, respectively), regardless of the type of ECMO. This drop in platelet count was associated with a decrease in platelet GP Ibα expression (before: 47.8±9.1 versus 24 hours post-ECMO: 42.3±8.9 mean fluorescence intensity; P=0.002) and an increase in soluble GP Ibα plasma levels (before: 5.6±3.3 versus 24 hours post-ECMO: 10.8±4.1 µg/mL; P<0.0001). GP Ibα shedding was also observed ex vivo and was unaffected by (1) red blood cells, (2) the coagulation potential, (3) an antibody blocking VWF-GP Ibα interaction, (4) an antibody limiting VWF degradation, and (5) supraphysiological VWF plasma concentrations. In contrast, GP Ibα shedding was dependent on rheological conditions, with a 2.8-fold increase at high- versus low-flow rates. Platelets perfused at high-flow rates before being transfused to immunodeficient mice were eliminated faster in vivo with an accelerated clearance of GP Ibα-negative versus GP Ibα-positive platelets. CONCLUSIONS ECMO-associated shear forces induce GP Ibα shedding and thrombocytopenia due to faster clearance of GP Ibα-negative platelets. Inhibiting GP Ibα shedding could represent an approach to reduce thrombocytopenia during ECMO.
Collapse
Affiliation(s)
- Antoine Rauch
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
- Department of Hematology and Transfusion, UFR3S-Université de Lille (A.R., A.D., M.D., E..J., M.D., S.S.)
| | - Annabelle Dupont
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
- Department of Hematology and Transfusion, UFR3S-Université de Lille (A.R., A.D., M.D., E..J., M.D., S.S.)
| | - Mickael Rosa
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
- Department of Hematology and Transfusion, UFR3S-Université de Lille (A.R., A.D., M.D., E..J., M.D., S.S.)
| | - Maximilien Desvages
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
| | - Christina Le Tanno
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
| | - Johan Abdoul
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
| | - Mélusine Didelot
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
| | - Alexandre Ung
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
| | - Richard Ruez
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
| | - Emmanuelle Jeanpierre
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
- Department of Hematology and Transfusion, UFR3S-Université de Lille (A.R., A.D., M.D., E..J., M.D., S.S.)
| | - Mélanie Daniel
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
- Department of Hematology and Transfusion, UFR3S-Université de Lille (A.R., A.D., M.D., E..J., M.D., S.S.)
| | - Delphine Corseaux
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
| | - Hugues Spillemaeker
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
- Department of Cardiology, UFR3S-Université de Lille (H.S., F.V., E.V.B.)
| | - Julien Labreuche
- ULR 2694-METRICS: Évaluation des technologies de santé et des pratiques médicales (J.L.), CHU Lille, University Lille, France
| | - Bénédicte Pradines
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
| | - Natacha Rousse
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
- Department of Cardiac Surgery, UFR3S-Université de Lille (N.R., A.V.)
| | - Peter J Lenting
- INSERM, UMR-S 1176, Université Paris-Saclay, Le Kremlin Bicêtre, France (P.J.L., C.V.D., C.C.)
| | - Mouhamed D Moussa
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
| | - André Vincentelli
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
- Department of Cardiac Surgery, UFR3S-Université de Lille (N.R., A.V.)
| | | | - Bart Staels
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
| | - Flavien Vincent
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
- Department of Cardiology, UFR3S-Université de Lille (H.S., F.V., E.V.B.)
| | - Cécile V Denis
- INSERM, UMR-S 1176, Université Paris-Saclay, Le Kremlin Bicêtre, France (P.J.L., C.V.D., C.C.)
| | - Eric Van Belle
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
- Department of Cardiology, UFR3S-Université de Lille (H.S., F.V., E.V.B.)
| | - Caterina Casari
- INSERM, UMR-S 1176, Université Paris-Saclay, Le Kremlin Bicêtre, France (P.J.L., C.V.D., C.C.)
| | - Sophie Susen
- Inserm, Institut Pasteur de Lille, France (A.R., A.D., M.R., M. Desvages, C.L.T., J.A., M. Didelot, A.U., R.R., E.J., M. Daniel, D.C., H.S., B.P., N.R., M.D.M., A.V., B.S., F.V., E.V.B., S.S.), CHU Lille, University Lille, France
- Department of Hematology and Transfusion, UFR3S-Université de Lille (A.R., A.D., M.D., E..J., M.D., S.S.)
| |
Collapse
|
10
|
Hearn JI, Gardiner EE. Research and Clinical Approaches to Assess Platelet Function in Flowing Blood. Arterioscler Thromb Vasc Biol 2023; 43:1775-1783. [PMID: 37615110 DOI: 10.1161/atvbaha.123.317048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Platelet adhesion and activation is fundamental to the formation of a hemostatic response to limit loss of blood and instigate wound repair to seal a site of vascular injury. The process of platelet aggregate formation is supported by the coagulation system driving injury-proximal formation of thrombin, which converts fibrinogen to insoluble fibrin. This highly coordinated series of molecular and membranous events must be routinely achieved in flowing blood, at vascular fluid shear rates that place significant strain on molecular and cellular interactions. Platelets have long been recognized to be able to slow down and adhere to sites of vascular injury and then activate and recruit more platelets that forge and strengthen adhesive ties with the vascular wall under these conditions. It has been a major challenge for the Platelet Research Community to construct experimental conditions that allow precise definition of the molecular steps occurring under flow. This brief review will discuss work to date from our group, as well as others that has furthered our understanding of platelet function in flowing blood.
Collapse
Affiliation(s)
- James I Hearn
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Elizabeth E Gardiner
- Division of Genome Science and Cancer, John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| |
Collapse
|
11
|
Martyanov AA, Tesakov IP, Khachatryan LA, An OI, Boldova AE, Ignatova AA, Koltsova EM, Korobkin JJD, Podoplelova NA, Svidelskaya GS, Yushkova E, Novichkova GA, Eble JA, Panteleev MA, Kalinin DV, Sveshnikova AN. Platelet functional abnormalities in pediatric patients with kaposiform hemangioendothelioma/Kasabach-Merritt phenomenon. Blood Adv 2023; 7:4936-4949. [PMID: 37307200 PMCID: PMC10463204 DOI: 10.1182/bloodadvances.2022009590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 05/05/2023] [Accepted: 05/21/2023] [Indexed: 06/14/2023] Open
Abstract
Kaposiform hemangioendothelioma (KHE) is a rare vascular tumor of infancy that is commonly associated with a life-threatening thrombocytopenic condition, Kasabach-Merritt phenomenon (KMP). Platelet CLEC-2, tumor podoplanin interaction is considered the key mechanism of platelet clearance in these patients. Here, we aimed to assess platelet functionality in such patients. Three groups of 6 to 9 children were enrolled: group A with KHE/KMP without hematologic response (HR) to therapy; group B with KHE/KMP with HR; and group C with healthy children. Platelet functionality was assessed by continuous and end point flow cytometry, low-angle light scattering analysis (LaSca), fluorescent microscopy of blood smears, and ex vivo thrombi formation. Platelet integrin activation in response to a combination of CRP (GPVI agonist) and TRAP-6 (PAR1 agonist), as well as calcium mobilization and integrin activation in response to CRP or rhodocytin (CLEC-2 agonist) alone, were significantly diminished in groups A and B. At the same time, platelet responses to ADP with or without TRAP-6 were unaltered. Thrombi formation from collagen in parallel plate flow chambers was also noticeably decreased in groups A and B. In silico analysis of these results predicted diminished amounts of CLEC-2 on the platelet surface of patients, which was further confirmed by immunofluorescence microscopy and flow cytometry. In addition, we also noted a decrease in GPVI levels on platelets from group A. In KHE/KMP, platelet responses induced by CLEC-2 or GPVI activation are impaired because of the diminished number of receptors on the platelet surface. This impairment correlates with the severity of the disease and resolves as the patient recovers.
Collapse
Affiliation(s)
- Alexey A. Martyanov
- Dmitry Rogachev National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Ivan P. Tesakov
- Dmitry Rogachev National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Lili A. Khachatryan
- Dmitry Rogachev National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Olga I. An
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
| | - Anna E. Boldova
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
| | - Anastasia A. Ignatova
- Dmitry Rogachev National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
| | - Ekaterina M. Koltsova
- Dmitry Rogachev National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
| | - Julia-Jessica D. Korobkin
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
| | - Nadezhda A. Podoplelova
- Dmitry Rogachev National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
| | - Galina S. Svidelskaya
- Dmitry Rogachev National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
| | - Eugenia Yushkova
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
| | - Galina A. Novichkova
- Dmitry Rogachev National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Johannes A. Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Mikhail A. Panteleev
- Dmitry Rogachev National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - Dmitrii V. Kalinin
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| | - Anastasia N. Sveshnikova
- Dmitry Rogachev National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
12
|
Rodríguez CS, Charó N, Tatti S, Gómez RM, D’Atri LP, Schattner M. Regulation of megakaryo/thrombopoiesis by endosomal toll-like receptor 7 and 8 activation of CD34 + cells in a viral infection model. Res Pract Thromb Haemost 2023; 7:100184. [PMID: 37538496 PMCID: PMC10394566 DOI: 10.1016/j.rpth.2023.100184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND CD34+ cells, megakaryocytes (MKs), and platelets express toll-like receptors (TLRs) that enable these cells to amplify the host innate immune response. However, the role of TLR7/TLR8 activation in megakaryopoiesis has not yet been investigated. OBJECTIVES We evaluated the effect of coxsackievirus B3 (CVB3) and synthetic TLR7/TLR8 agonists on the development of human MKs and production of platelets. METHODS CD34+ cells from human umbilical cord were inoculated with CVB3 or stimulated with synthetic TLR7/TLR8 agonists and then cultured in the presence of thrombopoietin. RESULTS CD34+ cells, MK progenitor cells, and mature MKs expressed TLR7 and TLR8, and exposure to CVB3 resulted in productive infection, as determined by the presence of viral infectious particles in culture supernatants. Cell expansion, differentiation into MKs, MK maturation, and platelet biogenesis were significantly reduced in CD34+-infected cultures. The reduction in MK growth was not due to an alteration in cellular proliferation but was accompanied by an increase in cellular apoptosis and pyroptosis. Impairment of MK generation and maturation of viable cells were also associated with decreased expression of transcription factors involved in these processes. These effects were completely abrogated by TLR7 but not TLR8 antagonists and mimicked by TLR7 but not TLR8 agonists. CVB3 infection of CD34+ cells increased the immunophenotype of MKs characterized as CD148+/CD48+ or CD41+/CD53+ cells. CONCLUSION These data suggest a novel role of TLR7 in megakaryo/thrombopoiesis that may contribute to a better understanding of the molecular basis underlying thrombocytopenia and the immunologic role of MKs in viral infection processes.
Collapse
Affiliation(s)
- Camila Sofía Rodríguez
- Laboratory of Experimental Thrombosis and Immunobiology of Inflammation, IMEX-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Nancy Charó
- Laboratory of Experimental Thrombosis and Immunobiology of Inflammation, IMEX-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | | | - Ricardo Martín Gómez
- Laboratory of Animal Viruses, Institute of Biotechnology and Molecular Biology, UNLP-CONICET, La Plata, Argentina
| | - Lina Paola D’Atri
- Laboratory of Experimental Thrombosis and Immunobiology of Inflammation, IMEX-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis and Immunobiology of Inflammation, IMEX-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| |
Collapse
|
13
|
Kawano T, Hisada Y, Grover SP, Schug WJ, Paul DS, Bergmeier W, Mackman N. Decreased Platelet Reactivity and Function in a Mouse Model of Human Pancreatic Cancer. Thromb Haemost 2023; 123:501-509. [PMID: 36716775 PMCID: PMC10820933 DOI: 10.1055/s-0043-1761419] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cancer patients have increased thrombosis and bleeding compared with the general population. Cancer is associated with activation of both platelets and coagulation. Mouse models have been used to study the dysregulation of platelets and coagulation in cancer. We established a mouse model of pancreatic cancer in which tissue factor-expressing human pancreatic tumors (BxPC-3) are grown in nude mice. Tumor-bearing mice have an activated coagulation system and increased venous thrombosis compared to control mice. We also showed that tumor-derived, tissue factor-positive extracellular vesicles activated platelets ex vivo and in vivo. In this study, we determined the effect of tumors on a platelet-dependent arterial thrombosis model. Unexpectedly, we observed significantly reduced carotid artery thrombosis in tumor-bearing mice compared to controls. In addition, we observed significantly increased tail bleeding in tumor-bearing mice compared to controls. These results suggested that the presence of the tumor affected platelets. Indeed, tumor-bearing mice exhibited a significant decrease in platelet count and an increase in mean platelet volume and percentage of reticulated platelets, findings that are consistent with increased platelet turnover. Levels of the platelet activation marker platelet factor 4 were also increased in tumor-bearing mice. We also observed decreased platelet receptor expression in tumor-bearing mice and reduced levels of active αIIb/β3 integrin in response to PAR4 agonist peptide and convulxin in platelets from tumor-bearing mice compared with platelets from control mice. In summary, our study suggests that in tumor-bearing mice there is chronic platelet activation, leading to thrombocytopenia, decreased receptor expression, and impaired platelet adhesive function.
Collapse
Affiliation(s)
- Tomohiro Kawano
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Yohei Hisada
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Steven P. Grover
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Wyatt J. Schug
- Department of Biochemistry and Biophysics, UNC Blood Research Center, University of North Carolina at Chapel Hill, North Carolina, United States
| | - David S. Paul
- Department of Biochemistry and Biophysics, UNC Blood Research Center, University of North Carolina at Chapel Hill, North Carolina, United States
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, UNC Blood Research Center, University of North Carolina at Chapel Hill, North Carolina, United States
| | - Nigel Mackman
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
14
|
Trivigno SMG, Guidetti GF, Barbieri SS, Zarà M. Blood Platelets in Infection: The Multiple Roles of the Platelet Signalling Machinery. Int J Mol Sci 2023; 24:ijms24087462. [PMID: 37108623 PMCID: PMC10138547 DOI: 10.3390/ijms24087462] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Platelets are classically recognized for their important role in hemostasis and thrombosis but they are also involved in many other physiological and pathophysiological processes, including infection. Platelets are among the first cells recruited to sites of inflammation and infection and they exert their antimicrobial response actively cooperating with the immune system. This review aims to summarize the current knowledge on platelet receptor interaction with different types of pathogens and the consequent modulations of innate and adaptive immune responses.
Collapse
Affiliation(s)
- Silvia M G Trivigno
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
- University School for Advanced Studies, IUSS, 27100 Pavia, Italy
| | | | - Silvia Stella Barbieri
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy
| | - Marta Zarà
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy
| |
Collapse
|
15
|
de Alvarenga VG, Oliveira LS, Santos GO, Vivas-Ruiz DE, Borges MH, de Souza RCG, Eble JA, Moura-da-Silva AM, Sanchez EF. Rhomb-I, a P–I metalloproteinase from Lachesis muta rhombeata venom degrades vessel extra cellular matrix components and impairs platelet aggregation. Toxicon 2023; 228:107097. [PMID: 37028563 DOI: 10.1016/j.toxicon.2023.107097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 04/08/2023]
Abstract
Rhomb-I, a 23-kDa metalloproteinase was isolated from L. m. rhombeata venom. Its dimethylcasein proteolysis was abolished by metal chelators, and slightly enhanced by Ca2+ and Mg2+ ions, but inhibited by Co2+, Zn2+ and α2-macroglobulin. In aqueous solution, rhomb-I autoproteolyzed to a 20- and 11-kDa fragments at 37 °C. The amino acid sequence showed high homology with other snake venom metalloproteinases. Rhomb-I causes hemorrhage that may be ascribed to hydrolysis of essential basement membrane, extracellular matrix and plasma proteins. It preferentially cleaves the α-chains of fibrin (ogen). Rhomb-I inhibited convulxin- and von Willebrand factor (vWF)-induced aggregation on human platelets without significant effect on collagen-stimulated aggregation or other effectors. It digests vWF into a low-molecular-mass multimers of vWF and a rvWF-A1 domain to a 27-kDa fragment as revealed by western blotting with mouse anti-rvWF A1-domain IgG. Incubation of platelets with rhomb-I resulted in adhesion to and cleavage of platelet receptors glycoprotein (GP)Ibα and GPVI to release a 55-kDa soluble form. Both membrane glycoproteins GPIbα that binds vWF, together with GPVI which binds collagen, play a key role in mediating platelet adhesion/activation and can initiate (patho)physiological thrombus formation. Conclusions: rhomb-I is implicated in the pathophysiology of Lachesis envenoming by disrupting vasculature, hemostasis and platelet aggregation through impairing vWF-GPIb axis and blocking GPVI-collagen binding.
Collapse
Affiliation(s)
| | - Luciana S Oliveira
- Laboratório de Bioquímica de Proteínas de Venenos Animais, Fundação Ezequiel Dias, Belo Horizonte, Brazil
| | - Gustavo O Santos
- Laboratório de Bioquímica de Proteínas de Venenos Animais, Fundação Ezequiel Dias, Belo Horizonte, Brazil
| | - Dan E Vivas-Ruiz
- Laboratório de Biologia Molecular, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Márcia Helena Borges
- Laboratório de Proteômica e Aracnídeos, Fundação Ezequiel Dias, Belo Horizonte, Brazil
| | | | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Germany
| | | | - Eladio F Sanchez
- Laboratório de Bioquímica de Proteínas de Venenos Animais, Fundação Ezequiel Dias, Belo Horizonte, Brazil.
| |
Collapse
|
16
|
De Simone I, Baaten CCFMJ, Gibbins JM, Ten Cate H, Heemskerk JWM, Jones CI, van der Meijden PEJ. Repeated platelet activation and the potential of previously activated platelets to contribute to thrombus formation. JOURNAL OF THROMBOSIS AND HAEMOSTASIS : JTH 2023; 21:1289-1306. [PMID: 36754678 DOI: 10.1016/j.jtha.2023.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/16/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
BACKGROUND Especially in disease conditions, platelets can encounter activating agents in circulation. OBJECTIVES To investigate the extent to which previously activated platelets can be reactivated and whether in-and reactivation applies to different aspects of platelet activation and thrombus formation. METHODS Short-and long-term effects of glycoprotein VI (GPVI) and G protein-coupled receptor (GPCR) stimulation on platelet activation and aggregation potential were compared via flow cytometry and plate-based aggregation. Using fluorescence and electron microscopy, we assessed platelet morphology and content, as well as thrombus formation. RESULTS After 30 minutes of stimulation with thrombin receptor activator peptide 6 (TRAP6) or adenosine diphosphate (ADP), platelets secondarily decreased in PAC-1 binding and were less able to aggregate. The reversibility of platelets after thrombin stimulation was concentration dependent. Reactivation was possible via another receptor. In contrast, cross-linked collagen-related peptide (CRP-XL) or high thrombin stimulation evoked persistent effects in αIIbβ3 activation and platelet aggregation. However, after 60 minutes of CRP-XL or high thrombin stimulation, when αIIbβ3 activation slightly decreased, restimulation with ADP or CRP-XL, respectively, increased integrin activation again. Compatible with decreased integrin activation, platelet morphology was reversed. Interestingly, reactivation of reversed platelets again resulted in shape change and if not fully degranulated, additional secretion. Moreover, platelets that were previously activated with TRAP6 or ADP regained their potential to contribute to thrombus formation under flow. On the contrary, prior platelet triggering with CRP-XL was accompanied by prolonged platelet activity, leading to a decreased secondary platelet adhesion under flow. CONCLUSION This work emphasizes that prior platelet activation can be reversed, whereafter platelets can be reactivated through a different receptor. Reversed, previously activated platelets can contribute to thrombus formation.
Collapse
Affiliation(s)
- Ilaria De Simone
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands; Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK
| | - Constance C F M J Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands; Institute for Molecular Cardiovascular Research, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK
| | - Hugo Ten Cate
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands; Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands; Synapse Research Institute, Maastricht, the Netherlands
| | - Chris I Jones
- Institute for Cardiovascular and Metabolic Research, School of Biological Sciences, University of Reading, Reading, UK
| | - Paola E J van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands; Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center, Maastricht, the Netherlands.
| |
Collapse
|
17
|
Armstrong PC, Allan HE, Kirkby NS, Gutmann C, Joshi A, Crescente M, Mitchell JA, Mayr M, Warner TD. Temporal in vivo platelet labeling in mice reveals age-dependent receptor expression and conservation of specific mRNAs. Blood Adv 2022; 6:6028-6038. [PMID: 36037520 PMCID: PMC9699941 DOI: 10.1182/bloodadvances.2022007099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 08/18/2022] [Indexed: 12/14/2022] Open
Abstract
The proportion of young platelets, also known as newly formed or reticulated, within the overall platelet population has been clinically correlated with adverse cardiovascular outcomes. However, our understanding of this is incomplete because of limitations in the technical approaches available to study platelets of different ages. In this study, we have developed and validated an in vivo temporal labeling approach using injectable fluorescent antiplatelet antibodies to subdivide platelets by age and assess differences in functional and molecular characteristics. With this approach, we found that young platelets (<24 hours old) in comparison with older platelets respond to stimuli with greater calcium flux and degranulation and contribute more to the formation of thrombi in vitro and in vivo. Sequential sampling confirmed this altered functionality to be independent of platelet size, with distribution of sizes of tracked platelets commensurate with the global platelet population throughout their 5-day lifespan in the circulation. The age-associated decrease in thrombotic function was accompanied by significant decreases in the surface expression of GPVI and CD31 (PECAM-1) and an increase in CD9. Platelet messenger RNA (mRNA) content also decreased with age but at different rates for individual mRNAs indicating apparent conservation of those encoding granule proteins. Our pulse-chase-type approach to define circulating platelet age has allowed timely reexamination of commonly held beliefs regarding size and reactivity of young platelets while providing novel insights into the temporal regulation of receptor and protein expression. Overall, future application of this validated tool will inform age-based platelet heterogeneity in physiology and disease.
Collapse
Affiliation(s)
- Paul C. Armstrong
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Correspondence: Paul C. Armstrong, Centre for Immunobiology, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, 4 Newark St, London E1 2AT, United Kingdom;
| | - Harriet E. Allan
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Nicholas S. Kirkby
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London United Kingdom
| | - Clemens Gutmann
- King’s British Heart Foundation Centre, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Abhishek Joshi
- King’s British Heart Foundation Centre, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Marilena Crescente
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Jane A. Mitchell
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London United Kingdom
| | - Manuel Mayr
- King’s British Heart Foundation Centre, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Timothy D. Warner
- Centre for Immunobiology, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
18
|
Subtype-specific plasma signatures of platelet-related protein releasate in acute pulmonary embolism. Thromb Res 2022; 220:75-87. [DOI: 10.1016/j.thromres.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/21/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022]
|
19
|
Cheung HYF, Moran LA, Sickmann A, Heemskerk JWM, Garcia Á, Watson SP. Inhibition of Src but not Syk causes weak reversal of GPVI-mediated platelet aggregation measured by light transmission aggregometry. Platelets 2022; 33:1293-1300. [PMID: 35535424 DOI: 10.1080/09537104.2022.2069235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Src tyrosine kinases and spleen tyrosine kinase (Syk) have recently been shown to contribute to sustained platelet aggregation on collagen under arterial shear. In the present study, we have investigated whether Src and Syk are required for aggregation under minimal shear following activation of glycoprotein VI (GPVI) and have extended this to C-type lectin-like receptor-2 (CLEC-2) which signals through the same pathway. Aggregation was induced by the GPVI ligand collagen-related peptide (CRP) and the CLEC-2 ligand rhodocytin and monitored by light transmission aggregometry (LTA). Aggregation and tyrosine phosphorylation by both receptors were sustained for up to 50 min. The addition of inhibitors of Src, Syk or Bruton's tyrosine kinase (Btk) at 150 sec, by which time aggregation was maximal, induced rapid loss of tyrosine phosphorylation of their downstream proteins, but only Src kinase inhibition caused a weak (~10%) reversal in light transmission. A similar effect was observed when the inhibitors were combined with apyrase and indomethacin or glycoprotein IIb-IIIa (GPIIb-IIIa) antagonist, eptifibatide. On the other hand, activation of GPIIb-IIIa by GPVI in a diluted platelet suspension, as measured by binding of fluorescein isothiocyanate-labeled antibody specific for the activated GPIIb-IIIa (FITC-PAC1), was reversed on the addition of Src and Syk inhibitors showing that integrin activation is rapidly reversible in the absence of outside-in signals. The results demonstrate that Src but not Syk and Btk contribute to sustained aggregation as monitored by LTA, possibly as a result of inhibition of outside-in signaling from GPIIb-IIIa to the cytoskeleton through a Syk-independent pathway. This is in contrast to the role of Syk in supporting sustained aggregation on collagen under arterial shear.
Collapse
Affiliation(s)
- Hilaire Yam Fung Cheung
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Department of Bioanalytics, Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V, Dortmund, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Luis A Moran
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, and Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Albert Sickmann
- Department of Bioanalytics, Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V, Dortmund, Germany
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Ángel Garcia
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidad de Santiago de Compostela, and Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
20
|
Reversible Platelet Integrin αIIbβ3 Activation and Thrombus Instability. Int J Mol Sci 2022; 23:ijms232012512. [PMID: 36293367 PMCID: PMC9604507 DOI: 10.3390/ijms232012512] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/28/2022] Open
Abstract
Integrin αIIbβ3 activation is essential for platelet aggregation and, accordingly, for hemostasis and arterial thrombosis. The αIIbβ3 integrin is highly expressed on platelets and requires an activation step for binding to fibrinogen, fibrin or von Willebrand factor (VWF). A current model assumes that the process of integrin activation relies on actomyosin force-dependent molecular changes from a bent-closed and extended-closed to an extended-open conformation. In this paper we review the pathways that point to a functional reversibility of platelet αIIbβ3 activation and transient aggregation. Furthermore, we refer to mouse models indicating that genetic defects that lead to reversible platelet aggregation can also cause instable thrombus formation. We discuss the platelet agonists and signaling pathways that lead to a transient binding of ligands to integrin αIIbβ3. Our analysis points to the (autocrine) ADP P2Y1 and P2Y12 receptor signaling via phosphoinositide 3-kinases and Akt as principal pathways linked to reversible integrin activation. Downstream signaling events by protein kinase C, CalDAG-GEFI and Rap1b have not been linked to transient integrin activation. Insight into the functional reversibility of integrin activation pathways will help to better understand the effects of antiplatelet agents.
Collapse
|
21
|
Chen Y, Fu W, Zheng Y, Yang J, Liu Y, Qi Z, Wu M, Fan Z, Yin K, Chen Y, Gao W, Ding Z, Dong J, Li Q, Zhang S, Hu L. Galectin 3 enhances platelet aggregation and thrombosis via Dectin-1 activation: a translational study. Eur Heart J 2022; 43:3556-3574. [PMID: 35165707 PMCID: PMC9989600 DOI: 10.1093/eurheartj/ehac034] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/25/2021] [Accepted: 01/18/2022] [Indexed: 01/25/2023] Open
Abstract
AIMS Galectin-3, a β-galactoside-binding lectin, is abnormally increased in cardiovascular disease. Plasma Galectin-3 receives a Class II recommendation for heart failure management and has been extensively studied for multiple cellular functions. The direct effects of Galectin-3 on platelet activation remain unclear. This study explores the direct effects of Galectin-3 on platelet activation and thrombosis. METHODS AND RESULTS A strong positive correlation between plasma Galectin-3 concentration and platelet aggregation or whole blood thrombus formation was observed in patients with coronary artery disease (CAD). Multiple platelet function studies demonstrated that Galectin-3 directly potentiated platelet activation and in vivo thrombosis. Mechanistic studies using the Dectin-1 inhibitor, laminarin, and Dectin-1-/- mice revealed that Galectin-3 bound to and activated Dectin-1, a receptor not previously reported in platelets, to phosphorylate spleen tyrosine kinase and thus increased Ca2+ influx, protein kinase C activation, and reactive oxygen species production to regulate platelet hyperreactivity. TD139, a Galectin-3 inhibitor in a Phase II clinical trial, concentration dependently suppressed Galectin-3-potentiated platelet activation and inhibited occlusive thrombosis without exacerbating haemorrhage in ApoE-/- mice, which spontaneously developed increased plasma Galectin-3 levels. TD139 also suppressed microvascular thrombosis to protect the heart from myocardial ischaemia-reperfusion injury in ApoE-/- mice. CONCLUSION Galectin-3 is a novel positive regulator of platelet hyperreactivity and thrombus formation in CAD. As TD139 has potent antithrombotic effects without bleeding risk, Galectin-3 inhibitors may have therapeutic advantages as potential antiplatelet drugs for patients with high plasma Galectin-3 levels.
Collapse
Affiliation(s)
- Yufei Chen
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Wanrong Fu
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yunbo Zheng
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Yang
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yangyang Liu
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiyong Qi
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Meiling Wu
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT, USA
| | - Kanhua Yin
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yunfeng Chen
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Wen Gao
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhongren Ding
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianzeng Dong
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Li
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Si Zhang
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Liang Hu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
22
|
Xiang K, Yanshan H, Chunmei Z, Minmin G, Yan W, Xiaojia Y. GP5 regulates epithelial-mesenchymal transition in breast cancer via the PI3K/AKT signaling pathway. Exp Biol Med (Maywood) 2022; 247:1501-1517. [PMID: 35880886 PMCID: PMC9554163 DOI: 10.1177/15353702221110642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent evidences have shown that glycoprotein V (GP5) protein, which is initially considered as an important adhesion molecule unique to the megakaryocyte line, was also specifically expressed in malignant human breast epithelial cells. However, its expression level and function are not clear. This study aimed to reveal the abnormal expression of GP5 in breast cancer (BC), research the associations between the GP5 abnormal expression and BC progression, and explore the molecular mechanism of GP5 in BC. Immunohistochemistry, Western blot (WB), and quantitative reverse transcription-polymerase chain reaction (RT-PCR) assays were used to determine the expression patterns of GP5 in BC tissues and cells. The expression profiles of GP5 in the Cancer Genome Atlas databases were analyzed by UALCAN. The GP5 knockdown and over-expression BC cell lines were constructed and confirmed by RT-PCR and WB. Transcriptome sequencing and KEGG database were performed to screen cellular processes and signal pathways. Phosphatidylinositol 3-kinase (PI3K)/AKT pathway was verified by RT-qPCR, and epithelial-mesenchymal transition (EMT) was confirmed by WB. The results indicated GP5 was highly expressed in BC tissues and might play an important role as a cancer-promoting gene in BC. The high expression of GP5 was significantly associated with higher nuclear grade, higher TNM stage, and human epidermal growth factor receptor 2 (HER2) negativity. GP5 may promote the proliferation, invasion, and metastasis of BC cells by activating PI3K/AKT signaling pathway to up-regulate the EMT. This study provides a new idea that GP5 was expected to become a potential molecular target for early BC clinic diagnosis and treatment.
Collapse
Affiliation(s)
- Kui Xiang
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China
| | - Hua Yanshan
- Department of Pathology, The Third Affiliated Hospital of Kunming Medical University and Yunnan Cancer Center, Yunnan Cancer Hospital, Kunming 650118, China
| | - Zhao Chunmei
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China
| | - Guo Minmin
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China
| | - Wang Yan
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China,Wang Yan.
| | - Yi Xiaojia
- Department of Pathology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China
| |
Collapse
|
23
|
GPVI expression is linked to platelet size, age, and reactivity. Blood Adv 2022; 6:4162-4173. [PMID: 35561312 PMCID: PMC9327529 DOI: 10.1182/bloodadvances.2021006904] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 05/03/2022] [Indexed: 01/19/2023] Open
Abstract
Juvenile platelets show increased GPVI expression. These platelets are highly responsive and more abundant among large platelets.
Platelets within one individual display heterogeneity in reactivity, size, age, and expression of surface receptors. To investigate the combined intraindividual contribution of platelet size, platelet age, and receptor expression levels on the reactivity of platelets, we studied fractions of large and small platelets from healthy donors separated by using differential centrifugation. Size-separated platelet fractions were perfused over a collagen-coated surface to assess thrombus formation. Multicolor flow cytometry was used to characterize resting and stimulated platelet subpopulations, and platelet age was determined based on RNA and HLA-I labeling. Signal transduction was analyzed by measuring consecutive phosphorylation of serine/threonine-protein kinase Akt. Compared with small platelets, large platelets adhered faster to collagen under flow and formed larger thrombi. Among the large platelets, a highly reactive juvenile platelet subpopulation was identified with high glycoprotein VI (GPVI) expression. Elevated GPVI expression correlated with high HLA-I expression, RNA content, and increased platelet reactivity. There was a stronger difference in Akt phosphorylation and activation upon collagen stimulation between juvenile and older platelets than between large and small platelets. GPVI expression and platelet reactivity decreased throughout platelet storage at 22°C and was better maintained throughout cold storage at 4°C. We further detected higher GPVI expression in platelets of patients with immune thrombocytopenia. Our findings show that high GPVI expression is a feature of highly reactive juvenile platelets, which are predominantly found among the large platelet population, explaining the better performance of large platelets during thrombus formation. These data are important for studies of thrombus formation, platelet storage, and immune thrombocytopenia.
Collapse
|
24
|
Bendas G, Schlesinger M. The GPIb-IX complex on platelets: insight into its novel physiological functions affecting immune surveillance, hepatic thrombopoietin generation, platelet clearance and its relevance for cancer development and metastasis. Exp Hematol Oncol 2022; 11:19. [PMID: 35366951 PMCID: PMC8976409 DOI: 10.1186/s40164-022-00273-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/19/2022] [Indexed: 12/13/2022] Open
Abstract
The glycoprotein (GP) Ib-IX complex is a platelet receptor that mediates the initial interaction with subendothelial von Willebrand factor (VWF) causing platelet arrest at sites of vascular injury even under conditions of high shear. GPIb-IX dysfunction or deficiency is the reason for the rare but severe Bernard-Soulier syndrome (BSS), a congenital bleeding disorder. Although knowledge on GPIb-IX structure, its basic functions, ligands, and intracellular signaling cascades have been well established, several advances in GPIb-IX biology have been made in the recent years. Thus, two mechanosensitive domains and a trigger sequence in GPIb were characterized and its role as a thrombin receptor was deciphered. Furthermore, it became clear that GPIb-IX is involved in the regulation of platelet production, clearance and thrombopoietin secretion. GPIb is deemed to contribute to liver cancer development and metastasis. This review recapitulates these novel findings highlighting GPIb-IX in its multiple functions as a key for immune regulation, host defense, and liver cancer development.
Collapse
Affiliation(s)
- Gerd Bendas
- Department of Pharmacy, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Martin Schlesinger
- Department of Pharmacy, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121, Bonn, Germany. .,Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany.
| |
Collapse
|
25
|
Neagoe RAI, Gardiner EE, Stegner D, Nieswandt B, Watson SP, Poulter NS. Rac Inhibition Causes Impaired GPVI Signalling in Human Platelets through GPVI Shedding and Reduction in PLCγ2 Phosphorylation. Int J Mol Sci 2022; 23:3746. [PMID: 35409124 PMCID: PMC8998833 DOI: 10.3390/ijms23073746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/22/2022] [Accepted: 03/26/2022] [Indexed: 12/19/2022] Open
Abstract
Rac1 is a small Rho GTPase that is activated in platelets upon stimulation with various ligands, including collagen and thrombin, which are ligands for the glycoprotein VI (GPVI) receptor and the protease-activated receptors, respectively. Rac1-deficient murine platelets have impaired lamellipodia formation, aggregation, and reduced PLCγ2 activation, but not phosphorylation. The objective of our study is to investigate the role of Rac1 in GPVI-dependent human platelet activation and downstream signalling. Therefore, we used human platelets stimulated using GPVI agonists (collagen and collagen-related peptide) in the presence of the Rac1-specific inhibitor EHT1864 and analysed platelet activation, aggregation, spreading, protein phosphorylation, and GPVI clustering and shedding. We observed that in human platelets, the inhibition of Rac1 by EHT1864 had no significant effect on GPVI clustering on collagen fibres but decreased the ability of platelets to spread or aggregate in response to GPVI agonists. Additionally, in contrast to what was observed in murine Rac1-deficient platelets, EHT1864 enhanced GPVI shedding in platelets and reduced the phosphorylation levels of PLCγ2 following GPVI activation. In conclusion, Rac1 activity is required for both human and murine platelet activation in response to GPVI-ligands, but Rac1's mode of action differs between the two species.
Collapse
Affiliation(s)
- Raluca A. I. Neagoe
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (R.A.I.N.); (S.P.W.)
- Rudolf Virchow Centre, Institute of Experimental Biomedicine I, University Hospital Würzburg, University of Würzburg, 97080 Würzburg, Germany; (D.S.); (B.N.)
| | - Elizabeth E. Gardiner
- Division of Genome Science and Cancer, John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia;
| | - David Stegner
- Rudolf Virchow Centre, Institute of Experimental Biomedicine I, University Hospital Würzburg, University of Würzburg, 97080 Würzburg, Germany; (D.S.); (B.N.)
| | - Bernhard Nieswandt
- Rudolf Virchow Centre, Institute of Experimental Biomedicine I, University Hospital Würzburg, University of Würzburg, 97080 Würzburg, Germany; (D.S.); (B.N.)
| | - Steve P. Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (R.A.I.N.); (S.P.W.)
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Midlands B15 2TT, UK
| | - Natalie S. Poulter
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (R.A.I.N.); (S.P.W.)
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Midlands B15 2TT, UK
| |
Collapse
|
26
|
Al-Tamimi M, Qiao J, Gardiner EE. The utility of platelet activation biomarkers in thrombotic microangiopathies. Platelets 2022; 33:503-511. [PMID: 35287530 DOI: 10.1080/09537104.2022.2026912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Primary thrombotic microangiopathies (TMAs) are observed in thrombotic thrombocytopenic purpura (TTP) and hemolytic uremic syndrome (HUS), while secondary TMAs have a wide range of etiologies. Early diagnosis and treatment of TMA are critical for patient well-being; however, distinguishing TTP from HUS on presentation is particularly challenging. Thrombocytopenia and platelet activation are central to different types of TMAs, thus limiting the utility of standard diagnostic approaches to evaluate the platelet function and hemostatic capacity. Alternative means of quantifying and monitoring changes to platelet activation and function are urgently needed. Activated platelets have been shown to interact with proteins of the complement and coagulation cascades and form part of inflammation processes engaged in TMA. Increased levels of platelet surface receptors as well as increased plasma levels of platelet-derived soluble proteins have been reported in TMAs. Elevated levels of platelet-leukocyte aggregates and platelet microparticles are also reported in different types of TMAs. Larger prospective evaluations of platelet activation markers in TMA using standardized assays, with comparison to cohorts of patients with thrombosis, coagulopathy, and thrombocytopenia, to evaluate the clinical usefulness of platelet markers in TMA are now needed. This review will summarize the current knowledge around platelet activation markers and critically evaluate their utility in diagnosis and prognosis of TMA patients.
Collapse
Affiliation(s)
- Mohammad Al-Tamimi
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Elizabeth E Gardiner
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| |
Collapse
|
27
|
Tyagi T, Jain K, Gu SX, Qiu M, Gu VW, Melchinger H, Rinder H, Martin KA, Gardiner EE, Lee AI, Ho Tang W, Hwa J. A guide to molecular and functional investigations of platelets to bridge basic and clinical sciences. NATURE CARDIOVASCULAR RESEARCH 2022; 1:223-237. [PMID: 37502132 PMCID: PMC10373053 DOI: 10.1038/s44161-022-00021-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 01/17/2022] [Indexed: 07/29/2023]
Abstract
Platelets have been shown to be associated with pathophysiological process beyond thrombosis, demonstrating critical additional roles in homeostatic processes, such as immune regulation, and vascular remodeling. Platelets themselves can have multiple functional states and can communicate and regulate other cells including immune cells and vascular smooth muscle cells, to serve such diverse functions. Although traditional platelet functional assays are informative and reliable, they are limited in their ability to unravel platelet phenotypic heterogeneity and interactions. Developments in methods such as electron microscopy, flow cytometry, mass spectrometry, and 'omics' studies, have led to new insights. In this Review, we focus on advances in platelet biology and function, with an emphasis on current and promising methodologies. We also discuss technical and biological challenges in platelet investigations. Using coronavirus disease 2019 (COVID-19) as an example, we further describe the translational relevance of these approaches and the possible 'bench-to-bedside' utility in patient diagnosis and care.
Collapse
Affiliation(s)
- Tarun Tyagi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Kanika Jain
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Sean X Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale University School of Medicine, Yale New Haven Hospital, New Haven, CT, USA
| | - Miaoyun Qiu
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Vivian W Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Hannah Melchinger
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Henry Rinder
- Department of Laboratory Medicine, Yale University School of Medicine, Yale New Haven Hospital, New Haven, CT, USA
| | - Kathleen A Martin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| | - Elizabeth E Gardiner
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Alfred I Lee
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Wai Ho Tang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
28
|
Martyanov AA, Boldova AE, Stepanyan MG, An OI, Gur'ev AS, Kassina DV, Volkov AY, Balatskiy AV, Butylin AA, Karamzin SS, Filimonova EV, Tsarenko SV, Roumiantsev SA, Rumyantsev AG, Panteleev MA, Ataullakhanov FI, Sveshnikova AN. Longitudinal multiparametric characterization of platelet dysfunction in COVID-19: Effects of disease severity, anticoagulation therapy and inflammatory status. Thromb Res 2022; 211:27-37. [PMID: 35066204 PMCID: PMC8761024 DOI: 10.1016/j.thromres.2022.01.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/25/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022]
Affiliation(s)
- Alexey A Martyanov
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow 109029, Russia; National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow 117198, Russia; Institute for Biochemical Physics (IBCP), Russian Academy of Sciences (RAS), Moscow, Kosyigina 4, 119334, Russia
| | - Anna E Boldova
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow 109029, Russia
| | - Maria G Stepanyan
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow 109029, Russia; Faculty of Physics, Lomonosov Moscow State University, 1/2 Leninskie gory, Moscow 119991, Russia
| | - Olga I An
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow 109029, Russia; National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow 117198, Russia; Department of Normal Physiology, Sechenov First Moscow State Medical University, 8/2 Trubetskaya St., Moscow 119991, Russia
| | - Alexander S Gur'ev
- Moscow Regional Research and Clinical Institute (MONIKI), 61/2 Shchepkina ul., Moscow 129110, Russia; Medtechnopark Ltd., 8-2-383 Profsoyuznaya str., Moscow 117292, Russia
| | - Darya V Kassina
- Moscow Regional Research and Clinical Institute (MONIKI), 61/2 Shchepkina ul., Moscow 129110, Russia
| | - Alexey Y Volkov
- Medtechnopark Ltd., 8-2-383 Profsoyuznaya str., Moscow 117292, Russia
| | - Alexandr V Balatskiy
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, 27-1 Lomonosovski Prospekt, Moscow 119991, Russia
| | - Andrei A Butylin
- Faculty of Physics, Lomonosov Moscow State University, 1/2 Leninskie gory, Moscow 119991, Russia
| | - Sergei S Karamzin
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow 109029, Russia
| | | | | | - Sergei A Roumiantsev
- Central Clinical Hospital of Russian Academy of Science, Oktyabrsky 3, Troitsk, Moscow 108840, Russia
| | - Alexander G Rumyantsev
- National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow 117198, Russia
| | - Mikhail A Panteleev
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow 109029, Russia; National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow 117198, Russia; Faculty of Physics, Lomonosov Moscow State University, 1/2 Leninskie gory, Moscow 119991, Russia
| | - Fazoil I Ataullakhanov
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow 109029, Russia; National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow 117198, Russia; Faculty of Physics, Lomonosov Moscow State University, 1/2 Leninskie gory, Moscow 119991, Russia
| | - Anastasia N Sveshnikova
- Center for Theoretical Problems of Physico-Сhemical Pharmacology, Russian Academy of Sciences, 30 Srednyaya Kalitnikovskaya str., Moscow 109029, Russia; National Medical Research Centеr of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, 1 Samory Mashela St, Moscow 117198, Russia; Faculty of Physics, Lomonosov Moscow State University, 1/2 Leninskie gory, Moscow 119991, Russia; Department of Normal Physiology, Sechenov First Moscow State Medical University, 8/2 Trubetskaya St., Moscow 119991, Russia.
| |
Collapse
|
29
|
Crispin P, Hicks S, Coupland LA, Ali S, Gardiner EE. Cryoprecipitate as an alternative to platelet transfusion in thrombocytopenia. EJHAEM 2022; 3:80-85. [PMID: 35846213 PMCID: PMC9175722 DOI: 10.1002/jha2.358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/13/2021] [Accepted: 11/18/2021] [Indexed: 11/24/2022]
Abstract
Platelet transfusions are not always available for bleeding in severe thrombocytopenia, as storage outside of major centers is limited by their short shelf-life. Data are lacking to support alternative available blood products; however, additional fibrinogen has been shown to enhance clot formation in vitro. To test the hypothesis that cryoprecipitate supplementation could improve clot formation in severe thrombocytopenia, eight hematological malignancy patients with platelet counts under 10 × 109/L each had 10 units of apheresis cryoprecipitate transfused prior to planned prophylactic platelet transfusions. The primary endpoint of thromboelastometry amplitude at 20 min increased by a mean of 5.1 mm (p < 0.01) following cryoprecipitate transfusion despite persisting thrombocytopenia. Thromboelastometry clotting times reduced by a mean of 7.8 s (p < 0.05) and alpha angle increased by a mean of 10.6⁰ (p < 0.01). These results are consistent with cryoprecipitate enhancing the strength of the fibrin/platelet meshwork within the forming thrombus. While platelet transfusion remains the standard of care, where platelet supplies are limited, these data provide a rationale for the use of cryoprecipitate to obtain hemostasis in bleeding thrombocytopenic patients.
Collapse
Affiliation(s)
- Philip Crispin
- ACRF Department of Cancer Biology and TherapeuticsThe John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralian Capital TerritoryAustralia
- Department of Clinical HaematologyCanberra HospitalGarranAustralian Capital TerritoryAustralia
| | - Sarah Hicks
- ACRF Department of Cancer Biology and TherapeuticsThe John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralian Capital TerritoryAustralia
| | - Lucy A. Coupland
- ACRF Department of Cancer Biology and TherapeuticsThe John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralian Capital TerritoryAustralia
| | - Sidra Ali
- ACRF Department of Cancer Biology and TherapeuticsThe John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralian Capital TerritoryAustralia
| | - Elizabeth E. Gardiner
- ACRF Department of Cancer Biology and TherapeuticsThe John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralian Capital TerritoryAustralia
| |
Collapse
|
30
|
Wang X, Wen C, Davis B, Shi P, Abune L, Lee K, Dong C, Wang Y. Synthetic DNA for Cell Surface Engineering: Experimental Comparison between Click Conjugation and Lipid Insertion in Terms of Cell Viability, Engineering Efficiency, and Displaying Stability. ACS APPLIED MATERIALS & INTERFACES 2022; 14:3900-3909. [PMID: 35020367 DOI: 10.1021/acsami.1c22774] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The cell surface can be engineered with synthetic DNA for various applications ranging from cancer immunotherapy to tissue engineering. However, while elegant methods such as click conjugation and lipid insertion have been developed to engineer the cell surface with DNA, little effort has been made to systematically evaluate and compare these methods. Resultantly, it is often challenging to choose a right method for a certain application or to interpret data from different studies. In this study, we systematically evaluated click conjugation and lipid insertion in terms of cell viability, engineering efficiency, and displaying stability. Cells engineered with both methods can maintain high viability when the concentration of modified DNA is less than 25-50 μM. However, lipid insertion is faster and more efficient in displaying DNA on the cell surface than click conjugation. The efficiency of displaying DNA with lipid insertion is 10-40 times higher than that with click conjugation for a large range of DNA concentration. However, the half-life of physically inserted DNA on the cell surface is 3-4 times lower than that of covalently conjugated DNA, which depends on the working temperature. While the half-life of physically inserted DNA molecules on the cell surface is shorter than that of DNA molecules clicked onto the cell surface, lipid insertion is more effective than click conjugation in the promotion of cell-cell interactions under the two different experimental settings. The data acquired in this work are expected to act as a guideline for choosing an approximate method for engineering the cell surface with synthetic DNA or even other biomolecules.
Collapse
Affiliation(s)
- Xuelin Wang
- Department of Biomedical Engineering, The Pennsylvania State University University Park, State College, Pennsylvania 16802, United States
| | - Connie Wen
- Department of Biomedical Engineering, The Pennsylvania State University University Park, State College, Pennsylvania 16802, United States
| | - Brandon Davis
- Department of Biomedical Engineering, The Pennsylvania State University University Park, State College, Pennsylvania 16802, United States
| | - Peng Shi
- Department of Biomedical Engineering, The Pennsylvania State University University Park, State College, Pennsylvania 16802, United States
| | - Lidya Abune
- Department of Biomedical Engineering, The Pennsylvania State University University Park, State College, Pennsylvania 16802, United States
| | - Kyungsene Lee
- Department of Biomedical Engineering, The Pennsylvania State University University Park, State College, Pennsylvania 16802, United States
| | - Cheng Dong
- Department of Biomedical Engineering, The Pennsylvania State University University Park, State College, Pennsylvania 16802, United States
| | - Yong Wang
- Department of Biomedical Engineering, The Pennsylvania State University University Park, State College, Pennsylvania 16802, United States
| |
Collapse
|
31
|
Baidildinova G, Nagy M, Jurk K, Wild PS, Ten Cate H, van der Meijden PEJ. Soluble Platelet Release Factors as Biomarkers for Cardiovascular Disease. Front Cardiovasc Med 2021; 8:684920. [PMID: 34235190 PMCID: PMC8255615 DOI: 10.3389/fcvm.2021.684920] [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: 03/24/2021] [Accepted: 05/24/2021] [Indexed: 12/19/2022] Open
Abstract
Platelets are the main players in thrombotic diseases, where activated platelets not only mediate thrombus formation but also are involved in multiple interactions with vascular cells, inflammatory components, and the coagulation system. Although in vitro reactivity of platelets provides information on the function of circulating platelets, it is not a full reflection of the in vivo activation state, which may be relevant for thrombotic risk assessment in various disease conditions. Therefore, studying release markers of activated platelets in plasma is of interest. While this type of study has been done for decades, there are several new discoveries that highlight the need for a critical assessment of the available tests and indications for platelet release products. First, new insights have shown that platelets are not only prominent players in arterial vascular disease, but also in venous thromboembolism and atrial fibrillation. Second, knowledge of the platelet proteome has dramatically expanded over the past years, which contributed to an increasing array of tests for proteins released and shed from platelets upon activation. Identification of changes in the level of plasma biomarkers associated with upcoming thromboembolic events allows timely and individualized adjustment of the treatment strategy to prevent disease aggravation. Therefore, biomarkers of platelet activation may become a valuable instrument for acute event prognosis. In this narrative review based on a systematic search of the literature, we summarize the process of platelet activation and release products, discuss the clinical context in which platelet release products have been measured as well as the potential clinical relevance.
Collapse
Affiliation(s)
- Gaukhar Baidildinova
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands.,Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Magdolna Nagy
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site RhineMain, Mainz, Germany
| | - Philipp S Wild
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site RhineMain, Mainz, Germany.,Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Hugo Ten Cate
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands.,Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center, Maastricht, Netherlands
| | - Paola E J van der Meijden
- Departments of Biochemistry and Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands.,Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center, Maastricht, Netherlands
| |
Collapse
|
32
|
Loss of GPVI and GPIbα contributes to trauma-induced platelet dysfunction in severely injured patients. Blood Adv 2021; 4:2623-2630. [PMID: 32556282 DOI: 10.1182/bloodadvances.2020001776] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/17/2020] [Indexed: 11/20/2022] Open
Abstract
Trauma-induced coagulopathy (TIC) is a complex, multifactorial failure of hemostasis that occurs in 25% of severely injured patients and results in a fourfold higher mortality. However, the role of platelets in this state remains poorly understood. We set out to identify molecular changes that may underpin platelet dysfunction after major injury and to determine how they relate to coagulopathy and outcome. We performed a range of hemostatic and platelet-specific studies in blood samples obtained from critically injured patients within 2 hours of injury and collected prospective data on patient characteristics and clinical outcomes. We observed that, although platelet counts were preserved above critical levels, circulating platelets sampled from trauma patients exhibited a profoundly reduced response to both collagen and the selective glycoprotein VI (GPVI) agonist collagen-related peptide, compared with those from healthy volunteers. These responses correlated closely with overall clot strength and mortality. Surface expression of the collagen receptors GPIbα and GPVI was reduced on circulating platelets in trauma patients, with increased levels of the shed ectodomain fragment of GPVI detectable in plasma. Levels of shed GPVI were highest in patients with more severe injuries and TIC. Collectively, these observations demonstrate that platelets experience a loss of GPVI and GPIbα after severe injury and translate into a reduction in the responsiveness of platelets during active hemorrhage. In turn, they are associated with reduced hemostatic competence and increased mortality. Targeting proteolytic shedding of platelet receptors is a potential therapeutic strategy for maintaining hemostatic competence in bleeding and improving the efficacy of platelet transfusions.
Collapse
|
33
|
Aslan JE. Platelet Proteomes, Pathways, and Phenotypes as Informants of Vascular Wellness and Disease. Arterioscler Thromb Vasc Biol 2021; 41:999-1011. [PMID: 33441027 PMCID: PMC7980774 DOI: 10.1161/atvbaha.120.314647] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Platelets rapidly undergo responsive transitions in form and function to repair vascular endothelium and mediate hemostasis. In contrast, heterogeneous platelet subpopulations with a range of primed or refractory phenotypes gradually arise in chronic inflammatory and other conditions in a manner that may indicate or support disease. Qualitatively distinguishable platelet phenotypes are increasingly associated with a variety of physiological and pathological circumstances; however, the origins and significance of platelet phenotypic variation remain unclear and conceptually vague. As changes in platelet function in disease exhibit many similarities to platelets following the activation of platelet agonist receptors, the intracellular responses of platelets common to hemostasis and inflammation may provide insights to the molecular basis of platelet phenotype. Here, we review concepts around how protein-level relations-from platelet receptors through intracellular signaling events-may help to define platelet phenotypes in inflammation, immune responses, aging, and other conditions. We further discuss how representing systems-wide platelet proteomics data profiles as circuit-like networks of causally related intracellular events, or, pathway maps, may inform molecular definitions of platelet phenotype. In addition to offering insights into platelets as druggable targets, maps of causally arranged intracellular relations underlying platelet function can also advance precision and interceptive medicine efforts by leveraging platelets as accessible, dynamic, endogenous, circulating biomarkers of vascular wellness and disease. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Joseph E. Aslan
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Department of Chemical Physiology and Biochemistry and School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
34
|
Sanchez EF, Alvarenga VG, Oliveira LS, Oliveira DL, Estevao-Costa MI, Flores-Ortiz R, Eble JA. A fibrinolytic snake venom metalloproteinase, mutalysin-II, with antiplatelet activity and targeting capability toward glycoprotein GPIbα and glycoprotein GPVI. Biochimie 2021; 184:1-7. [PMID: 33548391 DOI: 10.1016/j.biochi.2021.01.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 12/12/2022]
Abstract
Glycoprotein (GP)Ib that binds von Willebrand factor (vWF) and glycoprotein (GP)VI, that binds collagen play a significant role in platelet activation and aggregation, and are potential targets for antithrombotic treatment. They are targeted by snake venom proteinases. The effect of a such proteinase, mutalysin-II, on platelet aggregation was examined using washed human platelets and platelet-rich plasma. Its proteolytic activity on vWF, on its binding partner GPIbα, and on GPVI was analyzed by SDS-PAGE, and immunodetection with the corresponding antibodies after blotting. Dose- and time-dependently, mutalysin-II inhibits aggregation of washed platelets induced by vWF plus ristocetin and by convulxin, but with no significant effect on platelet-rich-plasma. Furthermore, mutalysin-II cleaves vWF into low molecular mass multimers of vWF and a rvWF-A1 domain to realease a ∼27-kDa fragment detectable by SDS-PAGE and blotting with mouse anti-rvWF-A1-domain IgG. Moreover, GPVI was cut by mutalysin-II into a soluble ∼55-kDa ectodomain and a fragment of ∼35-kDa. Thus, mutalysin-II inhibits vWF-induced platelet aggregation via cleavage of bound vWF-A1, and its receptor GPIbα. The additional cleavage of, GPVI, blocks collagen-induced platelets. Our data highlight mutalysin-II as an interesting platelet-directed tool targeting vWF-GPIbα binding and particularly GPVI. Thus, it might be suited for antithrombotic therapy as its combined inactivation of two receptors does not significantly compromise hemostasis, but shows high efficacy and safety. Studies are needed to further develop and demonstrate its potential benefits.
Collapse
Affiliation(s)
- Eladio F Sanchez
- Research and Development Center, Ezequiel Dias Foundation, Belo Horizonte, MG, Brazil.
| | - Valeria G Alvarenga
- Research and Development Center, Ezequiel Dias Foundation, Belo Horizonte, MG, Brazil
| | - Luciana S Oliveira
- Research and Development Center, Ezequiel Dias Foundation, Belo Horizonte, MG, Brazil
| | - Débora L Oliveira
- Research and Development Center, Ezequiel Dias Foundation, Belo Horizonte, MG, Brazil
| | - Maria I Estevao-Costa
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Renzo Flores-Ortiz
- Center for Data and Knowledge Integration for Health, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, BA, Brazil
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| |
Collapse
|
35
|
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
|
36
|
Wu J, Heemskerk JWM, Baaten CCFMJ. Platelet Membrane Receptor Proteolysis: Implications for Platelet Function. Front Cardiovasc Med 2021; 7:608391. [PMID: 33490118 PMCID: PMC7820117 DOI: 10.3389/fcvm.2020.608391] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022] Open
Abstract
The activities of adhesion and signaling receptors in platelets are controlled by several mechanisms. An important way of regulation is provided by proteolytic cleavage of several of these receptors, leading to either a gain or a loss of platelet function. The proteases involved are of different origins and types: (i) present as precursor in plasma, (ii) secreted into the plasma by activated platelets or other blood cells, or (iii) intracellularly activated and cleaving cytosolic receptor domains. We provide a comprehensive overview of the proteases acting on the platelet membrane. We describe how these are activated, which are their target proteins, and how their proteolytic activity modulates platelet functions. The review focuses on coagulation-related proteases, plasmin, matrix metalloproteinases, ADAM(TS) isoforms, cathepsins, caspases, and calpains. We also describe how the proteolytic activities are determined by different platelet populations in a thrombus and conversely how proteolysis contributes to the formation of such populations.
Collapse
Affiliation(s)
- Jiayu Wu
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Johan W. M. Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Constance C. F. M. J. Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen, Germany
| |
Collapse
|
37
|
Yatsenko TA. POLYCLONAL ANTIBODIES AGAINST HUMAN PLASMINOGEN: PURIFICATION, CHARACTERIZATION AND APPLICATION. BIOTECHNOLOGIA ACTA 2020. [DOI: 10.15407/biotech13.06.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The plasminogen/plasmin system plays a crucial role in fibrinolysis and regulation of cell functions in a wide range of normal and pathological processes. Investigation of plasminogen/plasmin functions requires the availability of well-characterized and effective molecular tools, such as antibodies. In the present work, the isolation and characterization of rabbit polyclonal antibodies against human plasminogen are described and approaches for the identification of plasminogen and its fragments using the purified antibodies are demonstrated. For the antibodies isolation, standard animal immunization and blood collection procedures, serum isolation, protein salting out and affinity chromatography were performed. For the antibodies characterization and application, the following methods were used: enzyme linked immunoassay (ELISA), Western blotting, FITC-protein conjugation, flow cytometry and spectrofluorometry. The obtained polyclonal rabbit anti-human plasminogen antibodies interacted with human Glu- and Lys-plasminogen, kringles 1-3 and 1-4 of plasminogen, mini-plasminogen, the heavy and light chain of plasmin. We propose the application of anti-plasminogen antibodies for the direct ELISA, Western blot analysis, and for flow cytometry and spectrofluorometric analysis of plasminogen binding with cells. The obtained anti-plasminogen antibodies are promising tools for the investigation of plasminogen/plasmin system functions, either fibrinolytic or signaling.
Collapse
|
38
|
Platelets and Defective N-Glycosylation. Int J Mol Sci 2020; 21:ijms21165630. [PMID: 32781578 PMCID: PMC7460655 DOI: 10.3390/ijms21165630] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/04/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
N-glycans are covalently linked to an asparagine residue in a simple acceptor sequence of proteins, called a sequon. This modification is important for protein folding, enhancing thermodynamic stability, and decreasing abnormal protein aggregation within the endoplasmic reticulum (ER), for the lifetime and for the subcellular localization of proteins besides other functions. Hypoglycosylation is the hallmark of a group of rare genetic diseases called congenital disorders of glycosylation (CDG). These diseases are due to defects in glycan synthesis, processing, and attachment to proteins and lipids, thereby modifying signaling functions and metabolic pathways. Defects in N-glycosylation and O-glycosylation constitute the largest CDG groups. Clotting and anticlotting factor defects as well as a tendency to thrombosis or bleeding have been described in CDG patients. However, N-glycosylation of platelet proteins has been poorly investigated in CDG. In this review, we highlight normal and deficient N-glycosylation of platelet-derived molecules and discuss the involvement of platelets in the congenital disorders of N-glycosylation.
Collapse
|
39
|
He Q, Chen J, Yan J, Cai S, Xiong H, Liu Y, Peng D, Mo M, Liu Z. Tumor microenvironment responsive drug delivery systems. Asian J Pharm Sci 2020; 15:416-448. [PMID: 32952667 PMCID: PMC7486519 DOI: 10.1016/j.ajps.2019.08.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/30/2019] [Accepted: 08/21/2019] [Indexed: 12/12/2022] Open
Abstract
Conventional tumor-targeted drug delivery systems (DDSs) face challenges, such as unsatisfied systemic circulation, low targeting efficiency, poor tumoral penetration, and uncontrolled drug release. Recently, tumor cellular molecules-triggered DDSs have aroused great interests in addressing such dilemmas. With the introduction of several additional functionalities, the properties of these smart DDSs including size, surface charge and ligand exposure can response to different tumor microenvironments for a more efficient tumor targeting, and eventually achieve desired drug release for an optimized therapeutic efficiency. This review highlights the recent research progresses on smart tumor environment responsive drug delivery systems for targeted drug delivery. Dynamic targeting strategies and functional moieties sensitive to a variety of tumor cellular stimuli, including pH, glutathione, adenosine-triphosphate, reactive oxygen species, enzyme and inflammatory factors are summarized. Special emphasis of this review is placed on their responsive mechanisms, drug loading models, drawbacks and merits. Several typical multi-stimuli responsive DDSs are listed. And the main challenges and potential future development are discussed.
Collapse
Affiliation(s)
- Qunye He
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Jun Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Jianhua Yan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Shundong Cai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Hongjie Xiong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Yanfei Liu
- School of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Dongming Peng
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Miao Mo
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhenbao Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| |
Collapse
|
40
|
Montague SJ, Hicks SM, Lee CSM, Coupland LA, Parish CR, Lee WM, Andrews RK, Gardiner EE. Fibrin exposure triggers αIIbβ3-independent platelet aggregate formation, ADAM10 activity and glycoprotein VI shedding in a charge-dependent manner. J Thromb Haemost 2020; 18:1447-1458. [PMID: 32198957 DOI: 10.1111/jth.14797] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 03/11/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Collagen and fibrin engagement and activation of glycoprotein (GP) VI induces proteolytic cleavage of the GPVI ectodomain generating shed soluble GPVI (sGPVI). Collagen-mediated GPVI shedding requires intracellular signalling to release the sGPVI, mediated by A Disintegrin And Metalloproteinase 10 (ADAM10); however, the precise mechanism by which fibrin induces GPVI shedding remains elusive. Plasma sGPVI levels are elevated in patients with coagulopathies, sepsis, or inflammation and can predict onset of sepsis and sepsis-related mortality; therefore, it is clinically important to understand the mechanisms of GPVI shedding under conditions of minimal collagen exposure. OBJECTIVES Our aim was to characterize mechanisms by which fibrin-GPVI interactions trigger GPVI shedding. METHODS Platelet aggregometry, sGPVI ELISA, and an ADAM10 fluorescence resonance energy transfer assay were used to measure fibrin-mediated platelet responses. RESULTS Fibrin induced αIIbβ3-independent washed platelet aggregate formation, GPVI shedding, and increased ADAM10 activity, all of which were insensitive to pre-treatment with inhibitors of Src family kinases but were divalent cation- and metalloproteinase-dependent. In contrast, treatment of washed platelets with other GPVI ligands, collagen, and collagen-related peptide caused αIIbβ3-dependent platelet aggregation and GPVI release but did not increase constitutive ADAM10 activity. CONCLUSIONS Fibrin engages GPVI in a manner that differs from other GPVI ligands. Inclusion of polyanionic molecules disrupted fibrin-induced platelet aggregate formation and sGPVI release, suggesting that electrostatic charge may play a role in fibrin/GPVI engagement. It may be feasible to exploit this property and specifically disrupt GPVI/fibrin interactions whilst sparing GPVI/collagen engagement.Fibrin engages GPVI in a manner that differs from other GPVI ligands. Inclusion of polyanionic molecules disrupted fibrin-induced platelet aggregate formation and sGPVI release, suggesting that electrostatic charge may play a role in fibrin/GPVI engagement. It may be feasible to exploit this property and specifically disrupt GPVI/fibrin interactions whilst sparing GPVI/collagen engagement.
Collapse
Affiliation(s)
- Samantha J Montague
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Sarah M Hicks
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Christine S-M Lee
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Lucy A Coupland
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Christopher R Parish
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Woei M Lee
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
- Research School of Electrical, Energy and Materials Engineering, College of Engineering and Computer Science, The Australian National University, Canberra, ACT, Australia
| | - Robert K Andrews
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Elizabeth E Gardiner
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
41
|
Tiruppathi C, Regmi SC, Wang DM, Mo GCH, Toth PT, Vogel SM, Stan RV, Henkemeyer M, Minshall RD, Rehman J, Malik AB. EphB1 interaction with caveolin-1 in endothelial cells modulates caveolae biogenesis. Mol Biol Cell 2020; 31:1167-1182. [PMID: 32238105 PMCID: PMC7353165 DOI: 10.1091/mbc.e19-12-0713] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/24/2020] [Accepted: 03/25/2020] [Indexed: 12/16/2022] Open
Abstract
Caveolae, the cave-like structures abundant in endothelial cells (ECs), are important for multiple signaling processes such as production of nitric oxide and caveolae-mediated intracellular trafficking. Using superresolution microscopy, fluorescence resonance energy transfer, and biochemical analysis, we observed that the EphB1 receptor tyrosine kinase constitutively interacts with caveolin-1 (Cav-1), the key structural protein of caveolae. Activation of EphB1 with its ligand Ephrin B1 induced EphB1 phosphorylation and the uncoupling EphB1 from Cav-1 and thereby promoted phosphorylation of Cav-1 by Src. Deletion of Cav-1 scaffold domain binding (CSD) motif in EphB1 prevented EphB1 binding to Cav-1 as well as Src-dependent Cav-1 phosphorylation, indicating the importance of CSD in the interaction. We also observed that Cav-1 protein expression and caveolae numbers were markedly reduced in ECs from EphB1-deficient (EphB1-/-) mice. The loss of EphB1 binding to Cav-1 promoted Cav-1 ubiquitination and degradation, and hence the loss of Cav-1 was responsible for reducing the caveolae numbers. These studies identify the crucial role of EphB1/Cav-1 interaction in the biogenesis of caveolae and in coordinating the signaling function of Cav-1 in ECs.
Collapse
Affiliation(s)
- Chinnaswamy Tiruppathi
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
- The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612
| | - Sushil C. Regmi
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
| | - Dong-Mei Wang
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
| | - Gary C. H. Mo
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
| | - Peter T. Toth
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
| | - Stephen M. Vogel
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
| | - Radu V. Stan
- Department of Pathology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755
| | - Mark Henkemeyer
- Departments of Neuroscience and Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Richard D. Minshall
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
- Anesthesiology, The University of Illinois College of Medicine, Chicago, IL 60612
- The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612
| | - Jalees Rehman
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
| | - Asrar B. Malik
- Departments of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612
- The Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, IL 60612
| |
Collapse
|
42
|
Waters L, Padula MP, Marks DC, Johnson L. Calcium chelation: a novel approach to reduce cryopreservation-induced damage to frozen platelets. Transfusion 2020; 60:1552-1563. [PMID: 32319689 DOI: 10.1111/trf.15799] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cryopreserved platelets are phenotypically and functionally different to conventionally stored platelets. Calcium may be released from internal stores during the freeze-thaw process, initiating signaling events which lead to these alterations. It was hypothesized that the addition of a calcium chelator prior to cryopreservation may mitigate some of these changes. METHODS Buffy coat-derived platelets that had been pooled and split were tested fresh and following cryopreservation (n = 8 per group). Platelets were cryopreserved using 5%-6% dimethylsulfoxide (DMSO) or were supplemented with increasing concentrations of the internal calcium chelator, BAPTA-AM (100 μM, 200 μM, or 400 μM), prior to storage at -80°C. RESULTS Supplementation of platelets with BAPTA-AM prior to freezing improved platelet recovery in a dose response manner (400 μM: 84 ± 2%) compared to standard DMSO cryopreserved platelets (70 ± 4%). There was a loss of GPIbα, GPVI, and GPIIb/IIIa receptors on platelets following cryopreservation, which was rescued when platelets were supplemented with BAPTA-AM (400 μM: p < 0.0001 for all). Platelet activation markers, such as phosphatidylserine and P-selectin, were externalized on platelets following cryopreservation. However, the addition of BAPTA-AM significantly reduced the increase of these activation markers on cryopreserved platelets (400 μM: p < 0.0001 for both). Both cryopreserved platelet groups exhibited similar functionality as assessed by thromboelastography, forming clots at a faster rate than fresh platelets. CONCLUSIONS This study demonstrates that calcium plays a crucial role in mediating cryopreservation-induced damage to frozen platelets. The addition of the calcium chelator, BAPTA-AM, prior to cryopreservation reduces this damage.
Collapse
Affiliation(s)
- Lauren Waters
- Research and Development, Australian Red Cross Lifeblood (formerly the Australian Red Cross Blood Service), Alexandria, New South Wales, Australia.,School of Life Sciences and Proteomics Core Facility, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Matthew P Padula
- School of Life Sciences and Proteomics Core Facility, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Denese C Marks
- Research and Development, Australian Red Cross Lifeblood (formerly the Australian Red Cross Blood Service), Alexandria, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Lacey Johnson
- Research and Development, Australian Red Cross Lifeblood (formerly the Australian Red Cross Blood Service), Alexandria, New South Wales, Australia
| |
Collapse
|
43
|
Maurer S, Kopp HG, Salih HR, Kropp KN. Modulation of Immune Responses by Platelet-Derived ADAM10. Front Immunol 2020; 11:44. [PMID: 32117229 PMCID: PMC7012935 DOI: 10.3389/fimmu.2020.00044] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/09/2020] [Indexed: 12/15/2022] Open
Abstract
Platelets have a crucial function in maintaining hemostasis. However, beyond their role in coagulation and thrombus formation, platelets have been implicated to affect various pathophysiological conditions such as infectious diseases, autoimmune disorders, and cancer. It is well-established that platelets aid local cancer growth by providing growth factors or contributing to cancer angiogenesis. In addition, they promote metastasis, among others by facilitation of tumor cell-extravasation and epithelial-to-mesenchymal-like transition as well as protecting metastasizing cancer cells from immunosurveillance. A variety of membrane-bound and soluble platelet-derived factors are involved in these processes, and many aspects of platelet biology in both health and disease are regulated by platelet-associated metalloproteinases and their inhibitors. Platelets synthesize (i) members of the matrix metalloproteinase (MMP) family and also inhibitors of MMPs such as members of the "tissue inhibitor of metalloproteinases" (TIMP) family as well as (ii) members of the "a disintegrin and metalloproteinase" (ADAM) family including ADAM10. Notably, platelet-associated metalloproteinase activity not only influences functions of platelets themselves: platelets can also induce expression and/or release of metalloproteinases e.g., in leukocytes or cancer cells, and ADAMs are emerging as important components by which platelets directly affect other cell types and function. This review outlines the function of metalloproteinases in platelet biology with a focus on ADAM10 and discusses the role of platelet-derived metalloproteinases in the interaction of platelets with components of the immune system and/or cancer cells.
Collapse
Affiliation(s)
- Stefanie Maurer
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany.,DFG Cluster of Excellence 2180 'Image-guided and Functional Instructed Tumor Therapy' (IFIT), University of Tuebingen, Tubingen, Germany.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Hans-Georg Kopp
- Departments of Molecular Oncology and Thoracic Oncology, Robert-Bosch-Hospital Stuttgart, Stuttgart, Germany
| | - Helmut R Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany.,DFG Cluster of Excellence 2180 'Image-guided and Functional Instructed Tumor Therapy' (IFIT), University of Tuebingen, Tubingen, Germany
| | - Korbinian N Kropp
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of Mainz, Mainz, Germany
| |
Collapse
|
44
|
Montague SJ, Lim YJ, Lee WM, Gardiner EE. Imaging Platelet Processes and Function-Current and Emerging Approaches for Imaging in vitro and in vivo. Front Immunol 2020; 11:78. [PMID: 32082328 PMCID: PMC7005007 DOI: 10.3389/fimmu.2020.00078] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 01/13/2020] [Indexed: 12/22/2022] Open
Abstract
Platelets are small anucleate cells that are essential for many biological processes including hemostasis, thrombosis, inflammation, innate immunity, tumor metastasis, and wound healing. Platelets circulate in the blood and in order to perform all of their biological roles, platelets must be able to arrest their movement at an appropriate site and time. Our knowledge of how platelets achieve this has expanded as our ability to visualize and quantify discreet platelet events has improved. Platelets are exquisitely sensitive to changes in blood flow parameters and so the visualization of rapid intricate platelet processes under conditions found in flowing blood provides a substantial challenge to the platelet imaging field. The platelet's size (~2 μm), rapid activation (milliseconds), and unsuitability for genetic manipulation, means that appropriate imaging tools are limited. However, with the application of modern imaging systems to study platelet function, our understanding of molecular events mediating platelet adhesion from a single-cell perspective, to platelet recruitment and activation, leading to thrombus (clot) formation has expanded dramatically. This review will discuss current platelet imaging techniques in vitro and in vivo, describing how the advancements in imaging have helped answer/expand on platelet biology with a particular focus on hemostasis. We will focus on platelet aggregation and thrombus formation, and how platelet imaging has enhanced our understanding of key events, highlighting the knowledge gained through the application of imaging modalities to experimental models in vitro and in vivo. Furthermore, we will review the limitations of current imaging techniques, and questions in thrombosis research that remain to be addressed. Finally, we will speculate how the same imaging advancements might be applied to the imaging of other vascular cell biological functions and visualization of dynamic cell-cell interactions.
Collapse
Affiliation(s)
- Samantha J. Montague
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Yean J. Lim
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
- Research School of Electrical, Energy and Materials Engineering, The Australian National University, Canberra, ACT, Australia
| | - Woei M. Lee
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
- Research School of Electrical, Energy and Materials Engineering, The Australian National University, Canberra, ACT, Australia
| | - Elizabeth E. Gardiner
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
45
|
Oliveira LS, Estevão-Costa MI, Alvarenga VG, Vivas-Ruiz DE, Yarleque A, Lima AM, Cavaco A, Eble JA, Sanchez EF. Atroxlysin-III, A Metalloproteinase from the Venom of the Peruvian Pit Viper Snake Bothrops atrox (Jergón) Induces Glycoprotein VI Shedding and Impairs Platelet Function. Molecules 2019; 24:molecules24193489. [PMID: 31561469 PMCID: PMC6803841 DOI: 10.3390/molecules24193489] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/28/2023] Open
Abstract
Atroxlysin-III (Atr-III) was purified from the venom of Bothrops atrox. This 56-kDa protein bears N-linked glycoconjugates and is a P-III hemorrhagic metalloproteinase. Its cDNA-deduced amino acid sequence reveals a multidomain structure including a proprotein, a metalloproteinase, a disintegrin-like and a cysteine-rich domain. Its identity with bothropasin and jararhagin from Bothrops jararaca is 97% and 95%, respectively. Its enzymatic activity is metal ion-dependent. The divalent cations, Mg2+ and Ca2+, enhance its activity, whereas excess Zn2+ inhibits it. Chemical modification of the Zn2+-complexing histidine residues within the active site by using diethylpyrocarbonate (DEPC) inactivates it. Atr-III degrades plasma fibronectin, type I-collagen, and mainly the α-chains of fibrinogen and fibrin. The von Willebrand factor (vWF) A1-domain, which harbors the binding site for GPIb, is not hydrolyzed. Platelets interact with collagen via receptors for collagen, glycoprotein VI (GPVI), and α2β1 integrin. Neither the α2β1 integrin nor its collagen-binding A-domain is fragmented by Atr-III. In contrast, Atr-III cleaves glycoprotein VI (GPVI) into a soluble ~55-kDa fragment (sGPVI). Thereby, it inhibits aggregation of platelets which had been stimulated by convulxin, a GPVI agonist. Selectively, Atr-III targets GPVI antagonistically and thus contributes to the antithrombotic effect of envenomation by Bothrops atrox.
Collapse
Affiliation(s)
- Luciana S Oliveira
- Research and Development Center, Ezequiel Dias Foundation, 30510-010 Belo Horizonte, MG, Brazil.
| | - Maria Inácia Estevão-Costa
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany.
| | - Valéria G Alvarenga
- Research and Development Center, Ezequiel Dias Foundation, 30510-010 Belo Horizonte, MG, Brazil.
| | - Dan E Vivas-Ruiz
- Laboratorio de Biología Molecular-Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Lima 14-0576, Peru.
| | - Armando Yarleque
- Laboratorio de Biología Molecular-Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Av. Venezuela Cdra 34 S/N, Ciudad Universitaria, Lima 01, Lima 14-0576, Peru.
| | - Augusto Martins Lima
- Laboratory of Hemodynamics and Cardiovascular Technology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.
| | - Ana Cavaco
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany.
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany.
| | - Eladio F Sanchez
- Research and Development Center, Ezequiel Dias Foundation, 30510-010 Belo Horizonte, MG, Brazil.
| |
Collapse
|
46
|
Zhang F, Liu Y, Lei J, Wang S, Ji X, Liu H, Yang Q. Metal-Organic-Framework-Derived Carbon Nanostructures for Site-Specific Dual-Modality Photothermal/Photodynamic Thrombus Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1901378. [PMID: 31508294 PMCID: PMC6724354 DOI: 10.1002/advs.201901378] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Indexed: 05/04/2023]
Abstract
Although near-infrared (NIR)-light-mediated photothermal thrombolysis has been investigated to overcome the bleeding risk of clinical clot-busting agents, the secondary embolism of post-phototherapy fragments (>10 µm) for small vessels should not be ignored in this process. In this study, dual-modality photothermal/photodynamic thrombolysis is explored using targeting nanoagents with an emphasis on improving biosafety as well as ameliorating the thrombolytic effect. The nanoagents can actively target glycoprotein IIb/IIIa receptors on thrombus to initiate site-specific thrombolysis by hyperthermia and reactive oxygen species under NIR laser irradiation. In comparison to single photothermal thrombolysis, an 87.9% higher re-establishment rate of dual-modality photothermal/photodynamic thrombolysis by one-time treatment is achieved in a lower limb thrombosis model. The dual-modality thrombolysis can also avoid re-embolization after breaking fibrin into tiny fragments. All the results show that this strategy is a safe and validated protocol for thrombolysis, which fits the clinical translational trend of nanomedicine.
Collapse
Affiliation(s)
- Fengrong Zhang
- Department of RadiologyXuanwu HospitalBeijing100053P. R. China
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringState Key Laboratory of Organic‐Inorganic CompositesBionanomaterials & Translational Engineering LaboratoryBeijing Key Laboratory of BioprocessBeijing Laboratory of Biomedical MaterialsBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Yuehong Liu
- Department of RadiologyXuanwu HospitalBeijing100053P. R. China
| | - Jiani Lei
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringState Key Laboratory of Organic‐Inorganic CompositesBionanomaterials & Translational Engineering LaboratoryBeijing Key Laboratory of BioprocessBeijing Laboratory of Biomedical MaterialsBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Shunhao Wang
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringState Key Laboratory of Organic‐Inorganic CompositesBionanomaterials & Translational Engineering LaboratoryBeijing Key Laboratory of BioprocessBeijing Laboratory of Biomedical MaterialsBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Xunming Ji
- Department of NeurosurgeryXuanwu HospitalBeijing100053P. R. China
| | - Huiyu Liu
- Beijing Advanced Innovation Center for Soft Matter Science and EngineeringState Key Laboratory of Organic‐Inorganic CompositesBionanomaterials & Translational Engineering LaboratoryBeijing Key Laboratory of BioprocessBeijing Laboratory of Biomedical MaterialsBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Qi Yang
- Department of RadiologyXuanwu HospitalBeijing100053P. R. China
| |
Collapse
|
47
|
Novel Stenotic Microchannels to Study Thrombus Formation in Shear Gradients: Influence of Shear Forces and Human Platelet-Related Factors. Int J Mol Sci 2019; 20:ijms20122967. [PMID: 31216638 PMCID: PMC6627598 DOI: 10.3390/ijms20122967] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/11/2019] [Accepted: 06/15/2019] [Indexed: 01/14/2023] Open
Abstract
Thrombus formation in hemostasis or thrombotic disease is initiated by the rapid adhesion, activation, and aggregation of circulating platelets in flowing blood. At arterial or pathological shear rates, for example due to vascular stenosis or circulatory support devices, platelets may be exposed to highly pulsatile blood flow, while even under constant flow platelets are exposed to pulsation due to thrombus growth or changes in vessel geometry. The aim of this study is to investigate platelet thrombus formation dynamics within flow conditions consisting of either constant or variable shear. Human platelets in anticoagulated whole blood were exposed ex vivo to collagen type I-coated microchannels subjected to constant shear in straight channels or variable shear gradients using different stenosis geometries (50%, 70%, and 90% by area). Base wall shears between 1800 and 6600 s−1, and peak wall shears of 3700 to 29,000 s−1 within stenoses were investigated, representing arterial-pathological shear conditions. Computational flow-field simulations and stenosis platelet thrombi total volume, average volume, and surface coverage were analysed. Interestingly, shear gradients dramatically changed platelet thrombi formation compared to constant base shear alone. Such shear gradients extended the range of shear at which thrombi were formed, that is, platelets became hyperthrombotic within shear gradients. Furthermore, individual healthy donors displayed quantifiable differences in extent/formation of thrombi within shear gradients, with implications for future development and testing of antiplatelet agents. In conclusion, here, we demonstrate a specific contribution of blood flow shear gradients to thrombus formation, and provide a novel platform for platelet functional testing under shear conditions.
Collapse
|
48
|
Nurden AT. Acquired Glanzmann thrombasthenia: From antibodies to anti-platelet drugs. Blood Rev 2019; 36:10-22. [PMID: 31010659 DOI: 10.1016/j.blre.2019.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/15/2019] [Accepted: 03/19/2019] [Indexed: 02/06/2023]
Abstract
In contrast to the inherited platelet disorder given by mutations in the ITGA2B and ITGB3 genes, mucocutaneous bleeding from a spontaneous inhibition of normally expressed αIIbβ3 characterizes acquired Glanzmann thrombasthenia (GT). Classically, it is associated with autoantibodies or paraproteins that block platelet aggregation without causing a fall in platelet count. However, inhibitory antibodies to αIIbβ3 are widely associated with primary immune thrombocytopenia (ITP), occur in secondary ITP associated with leukemia and related disorders, solid cancers and myeloma, other autoimmune diseases, following organ transplantation while cytoplasmic dysregulation of αIIbβ3 function features in myeloproliferative and myelodysplastic syndromes. Antibodies to αIIbβ3 occur during viral and bacterial infections, while drug-dependent antibodies reacting with αIIbβ3 are a special case. Direct induction of acquired GT is a feature of therapies that block platelets in coronary artery disease. This review looks at these conditions, emphasizing molecular mechanisms, therapy, patient management and future directions for research.
Collapse
Affiliation(s)
- Alan T Nurden
- Institut de Rhythmologie et de Modélisation Cardiaque, Plateforme Technologique d'Innovation Biomédicale, Hôpital Xavier Arnozan, Pessac, France.
| |
Collapse
|