1
|
Sawada S, Hitomi S, Hayashi Y, Yoshikawa K, Yagasaki F, Shinozuka H, Yonehara Y, Tsuboi Y, Iwata K, Shinoda M. P2Y 12 signaling in muscle satellite cells contributes to masseter muscle contraction-induced pain. THE JOURNAL OF PAIN 2025; 30:105360. [PMID: 40057215 DOI: 10.1016/j.jpain.2025.105360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/28/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
The mechanism behind masseter muscle pain, a major symptom of temporomandibular disorder (TMD), has remained poorly understood. Previous report indicates that adenosine triphosphate (ATP) is involved in the masseter muscle pain development, but the role of its hydrolysis product, adenosine diphosphate (ADP), remains uncertain. Consequently, this study aimed to elucidate the ADP role derived from the sustained masseter muscle contraction in the masseter muscle pain development. The right masseter muscle was electrically stimulated daily by placing electrodes on the muscle fascia, inducing strong contraction and mechanical allodynia. This led to an increment of the ATP release from the masseter muscle and a consequent increase in ADP produced by the hydrolysis of ATP. The mechanical allodynia was suppressed by intramuscular P2Y12 receptor antagonism and tumor necrosis factor alpha (TNF-α) inhibition. Additionally, muscle satellite cells expressed P2Y12 receptors, and the increase in amount of TNF-α released from these cells due to sustained contraction of the masseter muscle was suppressed by intramuscular P2Y12 receptor antagonism. These findings suggest that sustained masseter muscle contraction increases ADP levels within the muscle; this ADP, produced by the hydrolysis of ATP, promotes the release of TNF-α via P2Y12 receptors. The TNF-α signaling is likely to enhance the excitability of primary neurons projecting to the masseter muscle, thereby inducing masseter muscle pain. Therefore, it is plausible that TNF-α-induced nociceptive neuronal hyperexcitability through enhanced ADP signaling via P2Y12 receptors in satellite cells could be a candidate for therapeutic intervention for masseter muscle pain, a major symptom of TMD. PERSPECTIVE: Sustained masseter muscle contraction in rats induced mechanical allodynia and increased the amount of ADP within the muscle. Muscle satellite cells expressed P2Y12 receptors, and ADP-P2Y12 signaling increased the TNF-α release from these cells. TNF-α signaling enhanced the primary neuronal excitability, inducing masseter muscle pain.
Collapse
Affiliation(s)
- Sho Sawada
- Department of Oral and Maxillofacial Surgery Ⅱ, Nihon University School of Dentistry, Tokyo, Japan
| | - Suzuro Hitomi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Yoshinori Hayashi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Kenji Yoshikawa
- Department of Materials and Applied Chemistry, College of Science and Technology, Nihon University, Tokyo, Japan
| | - Fumitaka Yagasaki
- Department of Materials and Applied Chemistry, College of Science and Technology, Nihon University, Tokyo, Japan
| | - Hirotaka Shinozuka
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Yoshiyuki Yonehara
- Department of Oral and Maxillofacial Surgery Ⅱ, Nihon University School of Dentistry, Tokyo, Japan
| | - Yoshiyuki Tsuboi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Masamichi Shinoda
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan.
| |
Collapse
|
2
|
Kliuchnikov E, Peshkova AD, Vo MQ, Marx KA, Litvinov RI, Weisel JW, Purohit PK, Barsegov V. Exploring effects of platelet contractility on the kinetics, thermodynamics, and mechanisms of fibrin clot contraction. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:6. [PMID: 40012560 PMCID: PMC11850289 DOI: 10.1038/s44341-025-00011-9] [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: 09/10/2024] [Accepted: 01/22/2025] [Indexed: 02/28/2025]
Abstract
Mechanisms of blood clot contraction - platelet-driven fibrin network remodeling, are not fully understood. We developed a detailed computational ClotDynaMo model of fibrin network with activated platelets, whose clot contraction rate for normal 450,000/µl human platelets depends on serum viscosity η, platelet filopodia length l, and weakly depends on filopodia traction force f and filopodia extension-retraction speed v. Final clot volume is independent of η, but depends on v, f and l. Analysis of ClotDynaMo output revealed a 2.24 TJ/mol clot contraction free energy change, with ~67% entropy and ~33% internal energy changes. The results illuminate the "optimal contraction principle" that maximizes volume change while minimizing energy cost. An 8-chain continuum model of polymer elasticity containing platelet forces, captures clot contractility as a function of platelet count, η and l. The ClotDynaMo and continuum models can be extended to include red blood cells, variable platelet properties, and mechanics of fibrin network.
Collapse
Affiliation(s)
| | - Alina D. Peshkova
- Departments of Pharmacology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA USA
| | - Minh Quan Vo
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia, PA USA
| | - Kenneth A. Marx
- Department of Chemistry, University of Massachusetts, Lowell, MA USA
| | - Rustem I. Litvinov
- Departments of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA USA
| | - John W. Weisel
- Departments of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA USA
| | - Prashant K. Purohit
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia, PA USA
| | - Valeri Barsegov
- Department of Chemistry, University of Massachusetts, Lowell, MA USA
| |
Collapse
|
3
|
Tan YY, Liu J, Su QP. Advancing Platelet Research Through Live-Cell Imaging: Challenges, Techniques, and Insights. SENSORS (BASEL, SWITZERLAND) 2025; 25:491. [PMID: 39860861 PMCID: PMC11768609 DOI: 10.3390/s25020491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Platelet cells are essential to maintain haemostasis and play a critical role in thrombosis. They swiftly respond to vascular injury by adhering to damaged vessel surfaces, activating signalling pathways, and aggregating with each other to control bleeding. This dynamic process of platelet activation is intricately coordinated, spanning from membrane receptor maturation to intracellular interactions to whole-cell responses. Live-cell imaging has become an invaluable tool for dissecting these complexes. Despite its benefits, live imaging of platelets presents significant technical challenges. This review addresses these challenges, identifying key areas in need of further development and proposing possible solutions. We also focus on the dynamic processes of platelet adhesion, activation, and aggregation in haemostasis and thrombosis, applying imaging capacities from the microscale to the nanoscale. By exploring various live imaging techniques, we demonstrate how these approaches offer crucial insights into platelet biology and deepen our understanding of these three core events. In conclusion, this review provides an overview of the imaging methods currently available for studying platelet dynamics, guiding researchers in selecting suitable techniques for specific studies. By advancing our knowledge of platelet behaviour, these imaging methods contribute to research on haemostasis, thrombosis, and platelet-related diseases, ultimately aiming to improve clinical outcomes.
Collapse
Affiliation(s)
- Yuping Yolanda Tan
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia; (Y.Y.T.); (J.L.)
- Heart Research Institute, Newtown, NSW 2042, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Jinghan Liu
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia; (Y.Y.T.); (J.L.)
- Heart Research Institute, Newtown, NSW 2042, Australia
| | - Qian Peter Su
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia; (Y.Y.T.); (J.L.)
- Heart Research Institute, Newtown, NSW 2042, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
4
|
Zhao M, Peng D, Li Y, He M, Zhang Y, Zhou Q, Sun S, Ma P, Lv L, Wang X, Zhan L. Hemin regulates platelet clearance in hemolytic disease by binding to GPIbα. Platelets 2024; 35:2383642. [PMID: 39072582 DOI: 10.1080/09537104.2024.2383642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
Hemolysis is associated with thrombosis and vascular dysfunction, which are the pathological components of many diseases. Hemolytic products, including hemoglobin and hemin, activate platelets (PLT). Despite its activation, the effect of hemolysis on platelet clearance remains unclear, It is critical to maintain a normal platelet count and ensure that circulating platelets are functionally viable. In this study, we used hemin, a degradation product of hemoglobin, as a potent agonist to treat platelets and simulate changes in vivo in mice. Hemin treatment induced activation and morphological changes in platelets, including an increase in intracellular Ca2+ levels, phosphatidylserine (PS) exposure, and cytoskeletal rearrangement. Fewer hemin-treated platelets were cleared by macrophages in the liver after transfusion than untreated platelets. Hemin bound to glycoprotein Ibα (GPIbα), the surface receptor in hemin-induced platelet activation and aggregation. Furthermore, hemin decreased GPIbα desialylation, as evidenced by reduced Ricinus communis agglutinin I (RCA- I) binding, which likely extended the lifetime of such platelets in vivo. These data provided new insight into the mechanisms of GPIbα-mediated platelet activation and clearance in hemolytic disease.
Collapse
Affiliation(s)
- Man Zhao
- Field Blood Transfusion, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Dongxin Peng
- Field Blood Transfusion, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Yuxuan Li
- Field Blood Transfusion, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Minwei He
- Field Blood Transfusion, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Yulong Zhang
- Field Blood Transfusion, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Qianqian Zhou
- Field Blood Transfusion, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Sujing Sun
- Field Blood Transfusion, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Ping Ma
- Field Blood Transfusion, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Liping Lv
- Field Blood Transfusion, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Xiaohui Wang
- Field Blood Transfusion, Institute of Health Service and Transfusion Medicine, Beijing, China
| | - Linsheng Zhan
- Field Blood Transfusion, Institute of Health Service and Transfusion Medicine, Beijing, China
| |
Collapse
|
5
|
Khazali AS, Hadrawi WH, Ibrahim F, Othman S, Nor Rashid N. Thrombocytopenia in dengue infection: mechanisms and a potential application. Expert Rev Mol Med 2024; 26:e26. [PMID: 39397710 PMCID: PMC11488332 DOI: 10.1017/erm.2024.18] [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: 05/29/2023] [Revised: 03/18/2024] [Accepted: 05/30/2024] [Indexed: 10/15/2024]
Abstract
Thrombocytopenia is a common symptom and one of the warning signs of dengue virus (DENV) infection. Platelet depletion is critical as it may lead to other severe dengue symptoms. Understanding the molecular events of this condition during dengue infection is challenging because of the multifaceted factors involved in DENV infection and the dynamics of the disease progression. Platelet levels depend on the balance between platelet production and platelet consumption or clearance. Megakaryopoiesis and thrombopoiesis, two interdependent processes in platelet production, are hampered during dengue infection. Conversely, platelet elimination via platelet activation, apoptosis and clearance processes are elevated. Together, these anomalies contribute to thrombocytopenia in dengue patients. Targeting the molecular events of dengue-mediated thrombocytopenia shows great potential but still requires further investigation. Nonetheless, the application of new knowledge in this field, such as immature platelet fraction analysis, may facilitate physicians in monitoring the progression of the disease.
Collapse
Affiliation(s)
- Ahmad Suhail Khazali
- Faculty of Applied Sciences, Universiti Teknologi MARA (UiTM) Cawangan Perlis, Arau, Perlis, Malaysia
| | - Waqiyuddin Hilmi Hadrawi
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Fatimah Ibrahim
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
- Center for Innovation in Medical Engineering (CIME), Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Shatrah Othman
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Nurshamimi Nor Rashid
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Center for Innovation in Medical Engineering (CIME), Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
6
|
Sang Y, Lee RH, Luong A, Katona É, Whyte CS, Smith NL, Mast AE, Flick MJ, Mutch NJ, Bergmeier W, Wolberg AS. Activated platelets retain and protect most of their factor XIII-A cargo from proteolytic activation and degradation. Blood Adv 2024; 8:5072-5085. [PMID: 39116293 PMCID: PMC11459904 DOI: 10.1182/bloodadvances.2024012979] [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: 02/21/2024] [Revised: 07/25/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024] Open
Abstract
ABSTRACT Platelet factor XIII-A (FXIII-A) is a major cytoplasmic protein (∼3% of total), representing ∼50% of total circulating FXIII. However, mobilization of FXIII-A during platelet activation is not well defined. To determine mechanisms mediating the retention vs release of platelet FXIII-A, platelets from healthy humans and mice (F13a1-/-, Fga-/-, Plg-/-, Stim1fl/flPf4-Cre, and respective controls) were stimulated with thrombin, convulxin plus thrombin, or calcium ionophore (A23187), in the absence or presence of inhibitors of transglutaminase activity, messenger RNA (mRNA) translation, microtubule rearrangement, calpain, and Rho GTPase. Platelet releasates and pellets were separated by (ultra)centrifugation. FXIII-A was detected by immunoblotting and immunofluorescence microscopy. Even after strong dual agonist (convulxin plus thrombin) stimulation of human platelets, >80% platelet FXIII-A remained associated with the platelet pellet. In contrast, essentially all tissue factor pathway inhibitor, another cytoplasmic protein in platelets, was released to the supernatant. Pellet-associated FXIII-A was not due to de novo synthesis via platelet F13A1 mRNA. The proportion of platelet FXIII-A retained by vs released from activated platelets was partly dependent on STIM1 signaling, microtubule rearrangement, calpain, and RhoA activation but did not depend on the presence of fibrinogen or plasminogen. Immunofluorescence microscopy confirmed the presence of considerable FXIII-A within the activated platelets. Although released FXIII-A was cleaved to FXIII-A∗ and could be degraded by plasmin, platelet-associated FXIII-A remained uncleaved. Retention of substantial platelet-derived FXIII-A by activated platelets and its reduced susceptibility to thrombin- and plasmin-mediated proteolysis suggest platelet FXIII-A is a protected pool with biological role(s) that differs from plasma FXIII.
Collapse
Affiliation(s)
- Yaqiu Sang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC
- UNC Blood Research Center, Chapel Hill, NC
| | - Robert H. Lee
- UNC Blood Research Center, Chapel Hill, NC
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC
| | - Annie Luong
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC
- UNC Blood Research Center, Chapel Hill, NC
| | - Éva Katona
- Division of Clinical Laboratory Science, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Claire S. Whyte
- Aberdeen Cardiovascular & Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Nicholas L. Smith
- Department of Epidemiology, University of Washington, Seattle, WA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle, WA
| | - Alan E. Mast
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI
| | - Matthew J. Flick
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC
- UNC Blood Research Center, Chapel Hill, NC
| | - Nicola J. Mutch
- Aberdeen Cardiovascular & Diabetes Centre, Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Wolfgang Bergmeier
- UNC Blood Research Center, Chapel Hill, NC
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC
| | - Alisa S. Wolberg
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC
- UNC Blood Research Center, Chapel Hill, NC
| |
Collapse
|
7
|
Baxter RM, Harper MT. Dissecting the roles of dynamin and clathrin in platelet pinocytosis. Biochem Biophys Res Commun 2024; 725:150250. [PMID: 38870846 DOI: 10.1016/j.bbrc.2024.150250] [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: 05/16/2024] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
Platelets endocytose many molecules from their environment. However, this process of pinocytosis in platelets is poorly understood. Key endocytic regulators such as dynamin, clathrin, CDC42 and Arf6 are expressed in platelets but their roles in pinocytosis is not known. Stimulated platelets form two subpopulations of pro-aggregatory and procoagulant platelets. The effect of stimulation on pinocytosis is also poorly understood. In this study, washed human platelets were treated with a range of endocytosis inhibitors and stimulated using different activators. The rate of pinocytosis was assessed using pHrodo green, a pH-sensitive 10 kDa dextran. In unstimulated platelets, pHrodo fluorescence increased over time and accumulated as intracellular puncta indicating constituently active pinocytosis. Stimulated platelets (both pro-aggregatory and procoagulant) had an elevated pinocytosis rate compared to unstimulated platelets. Dynamin inhibition blocked pinocytosis in unstimulated, pro-aggregatory and procoagulant platelets indicating that most platelet pinocytosis is dynamin dependent. Although pinocytosis was clathrin-independent in unstimulated and procoagulant populations, clathrin partially contributed to pinocytosis in pro-aggregatory platelets.
Collapse
Affiliation(s)
- Ruby M Baxter
- Department of Pharmacology, University of Cambridge, United Kingdom
| | - Matthew T Harper
- Department of Pharmacology, University of Cambridge, United Kingdom.
| |
Collapse
|
8
|
Chernysh IN, Mukhopadhyay S, Johnson TA, Brooks JA, Sarkar R, Weisel JW, Antalis TM. Time-dependent ultrastructural changes during venous thrombogenesis and thrombus resolution. J Thromb Haemost 2024; 22:1675-1688. [PMID: 38492853 PMCID: PMC11139557 DOI: 10.1016/j.jtha.2024.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 02/08/2024] [Accepted: 02/21/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Deep vein thrombosis is a common vascular event that can result in debilitating morbidity and even death due to pulmonary embolism. Clinically, patients with faster resolution of a venous thrombus have improved prognosis, but the detailed structural information regarding changes that occur in a resolving thrombus over time is lacking. OBJECTIVES To define the spatial-morphologic characteristics of venous thrombus formation, propagation, and resolution at the submicron level over time. METHODS Using a murine model of stasis-induced deep vein thrombosis along with scanning electron microscopy and immunohistology, we determine the specific structural, compositional, and morphologic characteristics of venous thrombi formed after 4 days and identify the changes that take place during resolution by day 7. Comparison is made with the structure and composition of venous thrombi formed in mice genetically deficient in plasminogen activator inhibitor type 1. RESULTS As venous thrombus resolution progresses, fibrin exists in different structural forms, and there are dynamic cellular changes in the compositions of leukocytes, platelet aggregates, and red blood cells. Intrathrombus microvesicles are present that are not evident by histology, and red blood cells in the form of polyhedrocytes are an indicator of clot contraction. Structural evidence of fibrinolysis is observed early during thrombogenesis and is accelerated by plasminogen activator inhibitor type 1 deficiency. CONCLUSION The results reveal unique, detailed ultrastructural and compositional insights along with documentation of the dynamic changes that occur during accelerated resolution that are not evident by standard pathologic procedures and can be applied to inform diagnosis and effectiveness of thrombolytic treatments to improve patient outcomes.
Collapse
Affiliation(s)
- Irina N Chernysh
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Subhradip Mukhopadhyay
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Tierra A Johnson
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jacob A Brooks
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Rajabrata Sarkar
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - John W Weisel
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Toni M Antalis
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Research & Development Service, United States Department of Veterans Affairs Maryland Health Care System, Baltimore, Maryland, USA.
| |
Collapse
|
9
|
Pretorius E, Kell DB. A Perspective on How Fibrinaloid Microclots and Platelet Pathology May be Applied in Clinical Investigations. Semin Thromb Hemost 2024; 50:537-551. [PMID: 37748515 PMCID: PMC11105946 DOI: 10.1055/s-0043-1774796] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Microscopy imaging has enabled us to establish the presence of fibrin(ogen) amyloid (fibrinaloid) microclots in a range of chronic, inflammatory diseases. Microclots may also be induced by a variety of purified substances, often at very low concentrations. These molecules include bacterial inflammagens, serum amyloid A, and the S1 spike protein of severe acute respiratory syndrome coronavirus 2. Here, we explore which of the properties of these microclots might be used to contribute to differential clinical diagnoses and prognoses of the various diseases with which they may be associated. Such properties include distributions in their size and number before and after the addition of exogenous thrombin, their spectral properties, the diameter of the fibers of which they are made, their resistance to proteolysis by various proteases, their cross-seeding ability, and the concentration dependence of their ability to bind small molecules including fluorogenic amyloid stains. Measuring these microclot parameters, together with microscopy imaging itself, along with methodologies like proteomics and imaging flow cytometry, as well as more conventional assays such as those for cytokines, might open up the possibility of a much finer use of these microclot properties in generative methods for a future where personalized medicine will be standard procedures in all clotting pathology disease diagnoses.
Collapse
Affiliation(s)
- Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Matieland, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Douglas B. Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Matieland, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
10
|
Jourdi G, Ramström S, Sharma R, Bakchoul T, Lordkipanidzé M. Consensus report on flow cytometry for platelet function testing in thrombocytopenic patients: communication from the SSC of the ISTH. J Thromb Haemost 2023; 21:2941-2952. [PMID: 37481072 DOI: 10.1016/j.jtha.2023.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/24/2023]
Abstract
BACKGROUND Platelet count alone does not reliably predict bleeding risk, suggesting platelet function is important to monitor in patients with thrombocytopenia. There is still an unmet need for improved platelet function diagnostics in patients with low platelet count in many clinical situations. Flow cytometry is a promising tool allowing reliable platelet function study in this setting. OBJECTIVES The goal of this joint project between the International Society on Thrombosis and Haemostasis (ISTH) Scientific Standardization Committee (SSC) Subcommittees on Platelet Physiology and Platelet Immunology is to provide expert consensus guidance on the use of flow cytometry for the evaluation of platelet function, particularly activation, in patients with low platelet counts. METHODS A literature review was performed to identify relevant questions and areas of interest. An electronic expression of interest form was thereafter announced on the ISTH webpage, followed by a survey encompassing 37 issues regarding preanalytical, analytical, postanalytical, and performance aspects. Areas of disagreement or uncertainty were identified and formed the basis for 2 focus group discussions. RESULTS Consensus recommendations relative to patient sample collection, preanalytical variables, sample type, platelet-count cutoff, any potential specific modification of the standard flow cytometry protocol, and results expression and reporting are proposed based on the current practices of experts in the field as well as on literature review. CONCLUSION The proposed consensus recommendations would allow standardization of protocols in upcoming clinical studies. The clinical utility of platelet function testing using flow cytometry to predict bleeding risk still needs rigorous multicenter outcome studies in patients with thrombocytopenia.
Collapse
Affiliation(s)
- Georges Jourdi
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada; Université Paris Cité, INSERM, Innovative Therapies in Haemostasis, Paris, France; Service d'Hématologie Biologique, AP-HP, Hôpital Lariboisière, Paris, France
| | - Sofia Ramström
- Cardiovascular Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
| | - Ruchika Sharma
- Versiti Blood Center of Wisconsin Pediatric Hematology/Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Division of Hematology/Oncology/BMT, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Tamam Bakchoul
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tuebingen, University Hospital of Tuebingen, Tuebingen, Germany
| | - Marie Lordkipanidzé
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada; Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
11
|
Léopold V, Chouchane O, Butler JM, Schuurman AR, Michels EH, de Brabander J, Schomakers BV, van Weeghel M, Picavet-Havik DI, Grootemaat AE, Douma RA, Reijnders TD, Klarenbeek AM, Appelman B, Wiersinga WJ, van der Wel NN, den Dunnen J, Houtkooper RH, van't Veer C, van der Poll T. Platelets of COVID-19 patients display mitochondrial dysfunction, oxidative stress, and energy metabolism failure compatible with cell death. Res Pract Thromb Haemost 2023; 7:102213. [PMID: 38077825 PMCID: PMC10700394 DOI: 10.1016/j.rpth.2023.102213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Alterations in platelet function have been implicated in the pathophysiology of COVID-19 since the beginning of the pandemic. While early reports linked hyperactivated platelets to thromboembolic events in COVID-19, subsequent investigations demonstrated hyporeactive platelets with a procoagulant phenotype. Mitochondria are important for energy metabolism and the function of platelets. OBJECTIVES Here, we sought to map the energy metabolism of platelets in a cohort of noncritically ill COVID-19 patients and assess platelet mitochondrial function, activation status, and responsiveness to external stimuli. METHODS We enrolled hospitalized COVID-19 patients and controls between October 2020 and December 2021. Platelets function and metabolism was analyzed by flow cytometry, metabolomics, glucose fluxomics, electron and fluorescence microscopy and western blot. RESULTS Platelets from COVID-19 patients showed increased phosphatidylserine externalization indicating a procoagulant phenotype and hyporeactivity to ex vivo stimuli, associated with profound mitochondrial dysfunction characterized by mitochondrial depolarization, lower mitochondrial DNA-encoded transcript levels, an altered mitochondrial morphology consistent with increased mitochondrial fission, and increased pyruvate/lactate ratios in platelet supernatants. Metabolic profiling by untargeted metabolomics revealed NADH, NAD+, and ATP among the top decreased metabolites in patients' platelets, suggestive of energy metabolism failure. Consistently, platelet fluxomics analyses showed a strongly reduced utilization of 13C-glucose in all major energy pathways together with a rerouting of glucose to de novo generation of purine metabolites. Patients' platelets further showed evidence of oxidative stress, together with increased glutathione oxidation and synthesis. Addition of plasma from COVID-19 patients to normal platelets partially reproduced the phenotype of patients' platelets and disclosed a temporal relationship between mitochondrial decay and (subsequent) phosphatidylserine exposure and hyporeactivity. CONCLUSION These data link energy metabolism failure in platelets from COVID-19 patients with a prothrombotic platelet phenotype with features matching cell death.
Collapse
Affiliation(s)
- Valentine Léopold
- Amsterdam University Medical Center, University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Meibergdreef 9, Amsterdam, The Netherlands
- Université Paris Cité, INSERM UMR-S 942 (MASCOT), Paris, France
- Department of Anesthesiology and Critical Care and Burn Unit, Saint-Louis and Lariboisière Hospitals, Assistance Publique des Hôpitaux de Paris Nord, Paris, France
| | - Osoul Chouchane
- Amsterdam University Medical Center, University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Meibergdreef 9, Amsterdam, The Netherlands
| | - Joe M. Butler
- Amsterdam University Medical Center, University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Meibergdreef 9, Amsterdam, The Netherlands
| | - Alex R. Schuurman
- Amsterdam University Medical Center, University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Meibergdreef 9, Amsterdam, The Netherlands
| | - Erik H.A. Michels
- Amsterdam University Medical Center, University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Meibergdreef 9, Amsterdam, The Netherlands
| | - Justin de Brabander
- Amsterdam University Medical Center, University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Meibergdreef 9, Amsterdam, The Netherlands
| | - Bauke V. Schomakers
- Amsterdam University Medical Center, University of Amsterdam, Laboratory Genetic Metabolic Diseases, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism Institute, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam University Medical Center, University of Amsterdam, Core Facility Metabolomics, Meibergdreef 9, Amsterdam, The Netherlands
| | - Michel van Weeghel
- Amsterdam University Medical Center, University of Amsterdam, Laboratory Genetic Metabolic Diseases, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism Institute, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam University Medical Center, University of Amsterdam, Core Facility Metabolomics, Meibergdreef 9, Amsterdam, The Netherlands
| | - Daisy I. Picavet-Havik
- Amsterdam University Medical Center, University of Amsterdam, Electron Microscopy Centre Amsterdam, Medical Biology, Meibergdreef 9, Amsterdam, The Netherlands
| | - Anita E. Grootemaat
- Amsterdam University Medical Center, University of Amsterdam, Electron Microscopy Centre Amsterdam, Medical Biology, Meibergdreef 9, Amsterdam, The Netherlands
| | - Renée A. Douma
- Flevo Hospital, Department of Internal Medicine, Almere, The Netherlands
| | - Tom D.Y. Reijnders
- Amsterdam University Medical Center, University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Meibergdreef 9, Amsterdam, The Netherlands
| | - Augustijn M. Klarenbeek
- Amsterdam University Medical Center, University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Meibergdreef 9, Amsterdam, The Netherlands
| | - Brent Appelman
- Amsterdam University Medical Center, University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Meibergdreef 9, Amsterdam, The Netherlands
| | - W. Joost Wiersinga
- Amsterdam University Medical Center, University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam University Medical Center, University of Amsterdam, Division of Infectious Diseases, Meibergdreef 9, Amsterdam, The Netherlands
| | - Nicole N. van der Wel
- Amsterdam University Medical Center, University of Amsterdam, Electron Microscopy Centre Amsterdam, Medical Biology, Meibergdreef 9, Amsterdam, The Netherlands
| | - Jeroen den Dunnen
- Amsterdam University Medical Center, University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Meibergdreef 9, Amsterdam, The Netherlands
| | - Riekelt H. Houtkooper
- Amsterdam University Medical Center, University of Amsterdam, Laboratory Genetic Metabolic Diseases, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology and Metabolism Institute, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences Institute, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam University Medical Center, University of Amsterdam, Core Facility Metabolomics, Meibergdreef 9, Amsterdam, The Netherlands
| | - Cornelis van't Veer
- Amsterdam University Medical Center, University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Meibergdreef 9, Amsterdam, The Netherlands
| | - Tom van der Poll
- Amsterdam University Medical Center, University of Amsterdam, Center for Experimental and Molecular Medicine (CEMM), Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam University Medical Center, University of Amsterdam, Division of Infectious Diseases, Meibergdreef 9, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Ekhlak M, Kulkarni PP, Singh V, Chaurasia SN, Mohapatra SK, Chaurasia RN, Dash D. Necroptosis executioner MLKL plays pivotal roles in agonist-induced platelet prothrombotic responses and lytic cell death in a temporal order. Cell Death Differ 2023; 30:1886-1899. [PMID: 37301927 PMCID: PMC10406901 DOI: 10.1038/s41418-023-01181-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 05/04/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Necroptosis is a form of programmed cell death executed by receptor-interacting serine/threonine protein kinase 1 (RIPK1), RIPK3, and mixed lineage kinase domain-like (MLKL). Platelets are circulating cells that play central roles in haemostasis and pathological thrombosis. In this study we demonstrate seminal contribution of MLKL in transformation of agonist-stimulated platelets to active haemostatic units progressing eventually to necrotic death on a temporal scale, thus attributing a yet unrecognized fundamental role to MLKL in platelet biology. Physiological agonists like thrombin instigated phosphorylation and subsequent oligomerization of MLKL in platelets in a RIPK3-independent but phosphoinositide 3-kinase (PI3K)/AKT-dependent manner. Inhibition of MLKL significantly curbed agonist-induced haemostatic responses in platelets that included platelet aggregation, integrin activation, granule secretion, procoagulant surface generation, rise in intracellular calcium, shedding of extracellular vesicles, platelet-leukocyte interactions and thrombus formation under arterial shear. MLKL inhibition, too, prompted impairment in mitochondrial oxidative phosphorylation and aerobic glycolysis in stimulated platelets, accompanied with disruption in mitochondrial transmembrane potential, augmented proton leak and drop in both mitochondrial calcium as well as ROS. These findings underscore the key role of MLKL in sustaining OXPHOS and aerobic glycolysis that underlie energy-intensive platelet activation responses. Prolonged exposure to thrombin provoked oligomerization and translocation of MLKL to plasma membranes forming focal clusters that led to progressive membrane permeabilization and decline in platelet viability, which was prevented by inhibitors of PI3K/MLKL. In summary, MLKL plays vital role in transitioning of stimulated platelets from relatively quiescent cells to functionally/metabolically active prothrombotic units and their ensuing progression to necroptotic death.
Collapse
Affiliation(s)
- Mohammad Ekhlak
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Paresh P Kulkarni
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Vipin Singh
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Susheel N Chaurasia
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | | | - Rameshwar Nath Chaurasia
- Department of Neurology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Debabrata Dash
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
13
|
Peterson DF, McKibben NS, Hutchison CE, Lancaster K, Yang CJ, Dekeyser GJ, Friess DM, Schreiber MA, Willett NJ, Shatzel JJ, Aslan JE, Working ZM. Role of single-dose intravenous iron therapy for the treatment of anaemia after orthopaedic trauma: protocol for a pilot randomised controlled trial. BMJ Open 2023; 13:e069070. [PMID: 36944463 PMCID: PMC10032390 DOI: 10.1136/bmjopen-2022-069070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
INTRODUCTION Orthopaedic trauma and fracture care commonly cause perioperative anaemia and associated functional iron deficiency due to a systemic inflammatory state. Modern, strict transfusion thresholds leave many patients anaemic; managing this perioperative anaemia is an opportunity to impact outcomes in orthopaedic trauma surgery. The primary outcome of this pilot study is feasibility for a large randomised controlled trial (RCT) to evaluate intravenous iron therapy (IVIT) to improve patient well-being following orthopaedic injury. Measurements will include rate of participant enrolment, screening failure, follow-up, missing data, adverse events and protocol deviation. METHODS AND ANALYSIS This single-centre, pilot, double-blind RCT investigates the use of IVIT for acute blood loss anaemia in traumatically injured orthopaedic patients. Patients are randomised to receive either a single dose infusion of low-molecular weight iron dextran (1000 mg) or placebo (normal saline) postoperatively during their hospital stay for trauma management. Eligible subjects include adult patients admitted for lower extremity or pelvis operative fracture care with a haemoglobin of 7-11 g/dL within 7 days postoperatively during inpatient care. Exclusion criteria include history of intolerance to intravenous iron supplementation, active haemorrhage requiring ongoing blood product resuscitation, multiple planned procedures, pre-existing haematologic disorders or chronic inflammatory states, iron overload on screening or vulnerable populations. We follow patients for 3 months to measure the effect of iron supplementation on clinical outcomes (resolution of anaemia and functional iron deficiency), patient-reported outcomes (fatigue, physical function, depression and quality of life) and translational measures of immune cell function. ETHICS AND DISSEMINATION This study has ethics approval (Oregon Health & Science University Institutional Review Board, STUDY00022441). We will disseminate the findings through peer-reviewed publications and conference presentations. TRIAL REGISTRATION NUMBER NCT05292001; ClinicalTrials.gov.
Collapse
Affiliation(s)
- Danielle F Peterson
- Orthopaedics & Rehabilitation, Oregon Health & Science University, Portland, Oregon, USA
| | - Natasha S McKibben
- Orthopaedics & Rehabilitation, Oregon Health & Science University, Portland, Oregon, USA
| | - Catherine E Hutchison
- Orthopaedics & Rehabilitation, Oregon Health & Science University, Portland, Oregon, USA
| | - Karalynn Lancaster
- Orthopaedics & Rehabilitation, Oregon Health & Science University, Portland, Oregon, USA
| | - Chih Jen Yang
- Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Graham J Dekeyser
- Orthopaedics & Rehabilitation, Oregon Health & Science University, Portland, Oregon, USA
| | - Darin M Friess
- Orthopaedics & Rehabilitation, Oregon Health & Science University, Portland, Oregon, USA
| | - Martin A Schreiber
- Critical Care and Acute Care Surgery, Oregon Health & Science University, Portland, Oregon, USA
| | - Nick J Willett
- Bioengineering, University of Oregon, Eugene, Oregon, USA
| | - Joseph J Shatzel
- Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph E Aslan
- Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Zachary M Working
- Orthopaedics & Rehabilitation, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
14
|
NINJ1 Regulates Platelet Activation and PANoptosis in Septic Disseminated Intravascular Coagulation. Int J Mol Sci 2023; 24:ijms24044168. [PMID: 36835580 PMCID: PMC9958814 DOI: 10.3390/ijms24044168] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/12/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Disseminated intravascular coagulation (DIC), which is closely related to platelet activation, is a key factor leading to high mortality in sepsis. The release of contents from plasma membrane rupture after platelet death further aggravates thrombosis. Nerve injury-induced protein 1 (NINJ1) is a cell membrane protein that mediates membrane disruption, a typical marker of cell death, through oligomerization. Nevertheless, whether NINJ1 is expressed in platelets and regulates the platelet function remains unclear. The aim of this study was to evaluate the expression of NINJ1 in human and murine platelets and elucidate the role of NINJ1 in platelets and septic DIC. In this study, NINJ1 blocking peptide (NINJ126-37) was used to verify the effect of NINJ1 on platelets in vitro and in vivo. Platelet αIIbβ3 and P-selectin were detected by flow cytometry. Platelet aggregation was measured by turbidimetry. Platelet adhesion, spreading and NINJ1 oligomerization were examined by immunofluorescence. Cecal perforation-induced sepsis and FeCl3-induced thrombosis models were used to evaluate the role of NINJ1 in platelet, thrombus and DIC in vivo. We found that inhibition of NINJ1 alleviates platelet activation in vitro. The oligomerization of NINJ1 is verified in membrane-broken platelets, which is regulated by the PANoptosis pathway. In vivo studies demonstrate that inhibition of NINJ1 effectively reduces platelet activation and membrane disruption, thus suppressing platelet-cascade reaction and leading to anti-thrombosis and anti-DIC in sepsis. These data demonstrate that NINJ1 is critical in platelet activation and plasma membrane disruption, and inhibition of NINJ1 effectively reduces platelet-dependent thrombosis and DIC in sepsis. This is the first study to reveal the key role of NINJ1 in platelet and its related disorders.
Collapse
|
15
|
Hu JL, Zhang WJ. The role and pharmacological properties of P2Y12 receptor in cancer and cancer pain. Biomed Pharmacother 2023; 157:113927. [PMID: 36462316 DOI: 10.1016/j.biopha.2022.113927] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/22/2022] [Accepted: 10/24/2022] [Indexed: 12/05/2022] Open
Abstract
The G protein-coupled P2Y12 receptor (P2Y12R) was cloned in platelets and found to play a key role in maintaining platelet function in hemostasis and thrombosis, and these effects could be mediated by the P2Y12R. However, it has recently been found that P2Y12R-mediated the progression of tumor through interactions between platelets and tumor and stromal cells, as well as through products secreted by platelets. During tumor progression, tumor cells or other cells in the tumor microenvironment (such as immune cells) can secrete large amounts of ATP into the extracellular matrix, and extracellular ATP can be hydrolyzed into ADP. ADP is a P2Y12R activator and plays an important regulatory role in the proliferation and metastasis of tumor cells. P2Y12R is involved in platelet-cancer cell crosstalk and become a potential target for anticancer therapy. Moreover, tumor progression can induce pain, which seriously affects the quality of life of patients. P2Y12R is expressed in microglia and mediates the activities of microglial and participates in the occurrence of cancer pain. Conversely, inhibiting P2Y12R activation and down-regulating its expression has the effect of inhibiting tumor progression and pain. Therefore, P2Y12R can be a common therapeutic target for both. In this article, we explored the potential link between P2Y12R and cancer, discussed the intrinsic link of P2Y12R in cancer pain and the pharmacological properties of P2Y12R antagonists in the treatment of both.
Collapse
Affiliation(s)
- Jia-Ling Hu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province 343000, China
| | - Wen-Jun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi Province 343000, China.
| |
Collapse
|
16
|
Kim OV, Litvinov RI, Mordakhanova ER, Bi E, Vagin O, Weisel JW. Contribution of septins to human platelet structure and function. iScience 2022; 25:104654. [PMID: 35832887 PMCID: PMC9272382 DOI: 10.1016/j.isci.2022.104654] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 04/23/2022] [Accepted: 06/17/2022] [Indexed: 11/29/2022] Open
Abstract
Although septins have been well-studied in nucleated cells, their role in anucleate blood platelets remains obscure. Here, we elucidate the contribution of septins to human platelet structure and functionality. We show that Septin-2 and Septin-9 are predominantly distributed at the periphery of resting platelets and co-localize strongly with microtubules. Activation of platelets by thrombin causes clustering of septins and impairs their association with microtubules. Inhibition of septin dynamics with forchlorfenuron (FCF) reduces thrombin-induced densification of septins and lessens their colocalization with microtubules in resting and activated platelets. Exposure to FCF alters platelet shape, suggesting that septins stabilize platelet cytoskeleton. FCF suppresses platelet integrin αIIbβ3 activation, promotes phosphatidylserine exposure on activated platelets, and induces P-selectin expression on resting platelets, suggesting septin involvement in these processes. Inhibition of septin dynamics substantially reduces platelet contractility and abrogates their spreading on fibrinogen-coated surfaces. Overall, septins strongly contribute to platelet structure, activation and biomechanics.
Collapse
Affiliation(s)
- Oleg V. Kim
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rustem I. Litvinov
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elmira R. Mordakhanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Erfei Bi
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Olga Vagin
- Department of Pediatrics, Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, CA, USA
| | - John W. Weisel
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
17
|
Chronic Immune Platelet Activation Is Followed by Platelet Refractoriness and Impaired Contractility. Int J Mol Sci 2022; 23:ijms23137336. [PMID: 35806341 PMCID: PMC9266422 DOI: 10.3390/ijms23137336] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
Autoimmune diseases, including systemic lupus erythematosus (SLE), have a high risk of thrombotic and hemorrhagic complications associated with altered platelet functionality. We studied platelets from the blood of SLE patients and their reactivity. The surface expression of phosphatidylserine, P-selectin, and active integrin αIIbβ3 were measured using flow cytometry before and after platelet stimulation. Soluble P-selectin was measured in plasma. The kinetics of platelet-driven clot contraction was studied, as well as scanning and transmission electron microscopy of unstimulated platelets. Elevated levels of membrane-associated phosphatidylserine and platelet-attached and soluble P-selectin correlated directly with the titers of IgG, anti-dsDNA-antibodies, and circulating immune complexes. Morphologically, platelets in SLE lost their resting discoid shape, formed membrane protrusions and aggregates, and had a rough plasma membrane. The signs of platelet activation were associated paradoxically with reduced reactivity to a physiological stimulus and impaired contractility that revealed platelet exhaustion and refractoriness. Platelet activation has multiple pro-coagulant effects, and the inability to fully contract (retract) blood clots can be either a hemorrhagic or pro-thrombotic mechanism related to altered clot permeability, sensitivity of clots to fibrinolysis, obstructiveness, and embologenicity. Therefore, chronic immune platelet activation followed by secondary platelet dysfunction comprise an understudied pathogenic mechanism that supports hemostatic disorders in autoimmune diseases, such as SLE.
Collapse
|
18
|
Acquasaliente L, Pontarollo G, Radu CM, Peterle D, Artusi I, Pagotto A, Uliana F, Negro A, Simioni P, De Filippis V. Exogenous human α-Synuclein acts in vitro as a mild platelet antiaggregant inhibiting α-thrombin-induced platelet activation. Sci Rep 2022; 12:9880. [PMID: 35701444 PMCID: PMC9198058 DOI: 10.1038/s41598-022-12886-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 05/18/2022] [Indexed: 11/29/2022] Open
Abstract
α-Synuclein (αSyn) is a small disordered protein, highly conserved in vertebrates and involved in the pathogenesis of Parkinson’s disease (PD). Indeed, αSyn amyloid aggregates are present in the brain of patients with PD. Although the pathogenic role of αSyn is widely accepted, the physiological function of this protein remains elusive. Beyond the central nervous system, αSyn is expressed in hematopoietic tissue and blood, where platelets are a major cellular host of αSyn. Platelets play a key role in hemostasis and are potently activated by thrombin (αT) through the cleavage of protease-activated receptors. Furthermore, both αT and αSyn could be found in the same spatial environment, i.e. the platelet membrane, as αT binds to and activates platelets that can release αSyn from α-granules and microvesicles. Here, we investigated the possibility that exogenous αSyn could interfere with platelet activation induced by different agonists in vitro. Data obtained from distinct experimental techniques (i.e. multiple electrode aggregometry, rotational thromboelastometry, immunofluorescence microscopy, surface plasmon resonance, and steady-state fluorescence spectroscopy) on whole blood and platelet-rich plasma indicate that exogenous αSyn has mild platelet antiaggregating properties in vitro, acting as a negative regulator of αT-mediated platelet activation by preferentially inhibiting P-selectin expression on platelet surface. We have also shown that both exogenous and endogenous (i.e. cytoplasmic) αSyn preferentially bind to the outer surface of activated platelets. Starting from these findings, a coherent model of the antiplatelet function of αSyn is proposed.
Collapse
Affiliation(s)
- Laura Acquasaliente
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy
| | - Giulia Pontarollo
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy.,Center for Thrombosis and Hemostasis (CTH) University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Claudia Maria Radu
- Department of Women's & Children's Health, University of Padua, Padua, Italy.,Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University of Padua, via Giustiniani, 2, 35128, Padua, Italy
| | - Daniele Peterle
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy.,Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Ave. 02115, Boston, MA, USA
| | - Ilaria Artusi
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy
| | - Anna Pagotto
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy
| | - Federico Uliana
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy.,Institute of Molecular Systems Biology, ETH Zurich, 8093, Zurich, Switzerland
| | - Alessandro Negro
- Department of Biomedical Sciences, University of Padua, viale G. Colombo 3, 35100, Padua, Italy.
| | - Paolo Simioni
- Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University of Padua, via Giustiniani, 2, 35128, Padua, Italy.
| | - Vincenzo De Filippis
- Laboratory of Protein Chemistry and Molecular Hematology, Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padua, via Marzolo, 5, 35131, Padua, Italy. .,Biotechnology Center, CRIBI, University of Padua, Padua, Italy.
| |
Collapse
|
19
|
Elstrott BK, Lakshmanan HH, Melrose AR, Jordan KR, Martens KL, Yang C, Peterson DF, McMurry HS, Lavasseur C, Lo JO, Olson SR, DeLoughery TG, Aslan JE, Shatzel JJ. Platelet reactivity and platelet count in women with iron deficiency treated with intravenous iron. Res Pract Thromb Haemost 2022; 6:e12692. [PMID: 35356666 PMCID: PMC8941679 DOI: 10.1002/rth2.12692] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/31/2022] [Accepted: 02/18/2022] [Indexed: 12/03/2022] Open
Abstract
Background Iron deficiency anemia (IDA) and heavy menstrual bleeding are prevalent, interrelated issues impacting over 300 million premenopausal women worldwide. IDA is generally associated with increased platelet counts; however, the effects of IDA and its correction on platelet function in premenopausal women remain unknown. Objectives We sought to determine how IDA and intravenous iron affect platelet count and platelet function in premenopausal women. Methods Hematologic indices were assessed in a multicenter, retrospective cohort of 231 women repleted with intravenous iron. Pre- and postinfusion blood samples were then obtained from a prospective cohort of 13 women to analyze the effect of intravenous iron on hematologic parameters as well as platelet function with flow cytometry and platelet aggregation assays under physiologic shear. Results Following iron replacement, anemia improved, and mean platelet counts decreased by 26.5 and 16.0 K/mm3 in the retrospective and prospective cohorts, respectively. Replacement reduced baseline platelet surface P-selectin levels while enhancing platelet secretory responses to agonists, including collagen-related peptide and ADP. Platelet adhesion and aggregation on collagen under physiologic shear also significantly increased following repletion. Conclusion We find that intravenous iron improves anemia while restoring platelet counts and platelet secretory responses in premenopausal women with iron deficiency. Our results suggest that iron deficiency as well as iron replacement can have a range of effects on platelet production and function. Consequently, platelet reactivity profiles should be further examined in women and other groups with IDA where replacement offers a promising means to improve anemia as well as quality of life.
Collapse
Affiliation(s)
- Benjamin K. Elstrott
- Division of Hematology‐Medical OncologySchool of MedicineKnight Cancer InstituteOregon Health & Science UniversityPortlandOregonUSA
| | - Hari H.S. Lakshmanan
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandOregonUSA
| | - Alexander R. Melrose
- Division of CardiologySchool of MedicineKnight Cardiovascular InstituteOregon Health & Science UniversityPortlandOregonUSA
| | - Kelley R. Jordan
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandOregonUSA
| | - Kylee L. Martens
- Division of Hematology‐Medical OncologySchool of MedicineKnight Cancer InstituteOregon Health & Science UniversityPortlandOregonUSA
| | - Chih‐Jen Yang
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandOregonUSA
| | - Danielle F. Peterson
- Department of Orthopedics and RehabilitationOregon Health & Science UniversityPortlandOregonUSA
| | - Hannah Stowe McMurry
- Division of Hematology‐Medical OncologySchool of MedicineKnight Cancer InstituteOregon Health & Science UniversityPortlandOregonUSA
| | - Corinne Lavasseur
- Division of Hematology‐Medical OncologySchool of MedicineKnight Cancer InstituteOregon Health & Science UniversityPortlandOregonUSA
| | - Jamie O. Lo
- Department of Obstetrics and GynecologyOregon Health & Science UniversityPortlandOregonUSA
| | - Sven R. Olson
- Division of Hematology‐Medical OncologySchool of MedicineKnight Cancer InstituteOregon Health & Science UniversityPortlandOregonUSA
| | - Thomas G. DeLoughery
- Division of Hematology‐Medical OncologySchool of MedicineKnight Cancer InstituteOregon Health & Science UniversityPortlandOregonUSA
| | - Joseph E. Aslan
- Department of Biomedical EngineeringOregon Health & Science UniversityPortlandOregonUSA
- Division of CardiologySchool of MedicineKnight Cardiovascular InstituteOregon Health & Science UniversityPortlandOregonUSA
| | - Joseph J. Shatzel
- Division of Hematology‐Medical OncologySchool of MedicineKnight Cancer InstituteOregon Health & Science UniversityPortlandOregonUSA
- Division of CardiologySchool of MedicineKnight Cardiovascular InstituteOregon Health & Science UniversityPortlandOregonUSA
| |
Collapse
|
20
|
Extent of intravital contraction of arterial and venous thrombi and pulmonary emboli. Blood Adv 2021; 6:1708-1718. [PMID: 34972200 PMCID: PMC8941457 DOI: 10.1182/bloodadvances.2021005801] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/29/2021] [Indexed: 11/20/2022] Open
Abstract
Ratio of compressed polyhedral to native biconcave RBCs in blood clots and thrombi is a “ruler” to measure extent of clot contraction. The extent of intravital contraction of ex vivo arterial and venous thrombi is associated with their origins, age, and embologenicity.
Blood clots and thrombi undergo platelet-driven contraction/retraction followed by structural rearrangements. We have established quantitative relationships between the composition of blood clots and extent of contraction to determine intravital contraction of thrombi and emboli based on their content. The composition of human blood clots and thrombi was quantified using histology and scanning electron microscopy. Contracting blood clots were segregated into the gradually shrinking outer layer that contains a fibrin-platelet mesh and the expanding inner portion with compacted red blood cells (RBCs). At 10% contraction, biconcave RBCs were partially compressed into polyhedral RBCs, which became dominant at 20% contraction and higher. The polyhedral/biconcave RBC ratio and the extent of contraction displayed an exponential relationship, which was used to determine the extent of intravital contraction of ex vivo thrombi, ranging from 30% to 50%. In venous thrombi, the extent of contraction decreased gradually from the older (head) to the younger (body, tail) parts. In pulmonary emboli, the extent of contraction was significantly lower than in the venous head but was similar to the body and tail, suggesting that the emboli originate from the younger portion(s) of venous thrombi. The extent of contraction in arterial cerebral thrombi was significantly higher than in the younger parts of venous thrombi (body, tail) and pulmonary emboli but was indistinguishable from the older part (head). A novel tool, named the “contraction ruler,” has been developed to use the composition of ex vivo thrombi to assess the extent of their intravital contraction, which contributes to the pathophysiology of thromboembolism.
Collapse
|
21
|
Buijsers B, Garishah FM, Riswari SF, van Ast RM, Pramudo SG, Tunjungputri RN, Overheul GJ, van Rij RP, van der Ven A, Alisjahbana B, Gasem MH, de Mast Q, van der Vlag J. Increased Plasma Heparanase Activity and Endothelial Glycocalyx Degradation in Dengue Patients Is Associated With Plasma Leakage. Front Immunol 2021; 12:759570. [PMID: 34987504 PMCID: PMC8722520 DOI: 10.3389/fimmu.2021.759570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Background Endothelial hyper-permeability with plasma leakage and thrombocytopenia are predominant features of severe dengue virus infection. It is well established that heparanase, the endothelial glycocalyx degrading enzyme, plays a major role in various diseases with vascular leakage. It is yet to be elucidated whether heparanase activity plays a major role in dengue-associated plasma leakage. Moreover, the major source of heparanase secretion and activation in dengue remains elusive. Since a relatively high amount of heparanase is stored in platelets, we postulate that heparanase released by activated platelets contributes to the increased plasma heparanase activity during dengue virus infection. Methods Heparanase activity (plasma and urine), and heparan sulfate and syndecan-1 (plasma levels) were measured in dengue patients with thrombocytopenia in acute phase (n=30), during course of disease (n=10) and in convalescent phase (n=25). Associations with clinical parameters and plasma leakage markers were explored. Platelets from healthy donors were stimulated with dengue non-structural protein-1, DENV2 virus and thrombin to evaluate heparanase release and activity ex vivo. Results Heparanase activity was elevated in acute dengue and normalized during convalescence. Similarly, glycocalyx components, such as heparan sulfate and syndecan-1, were increased in acute dengue and restored during convalescence. Increased heparanase activity correlated with the endothelial dysfunction markers heparan sulfate and syndecan-1, as well as clinical markers of plasma leakage such as ascites, hematocrit concentration and gall-bladder wall thickening. Notably, platelet number inversely correlated with heparanase activity. Ex vivo incubation of platelets with thrombin and live DENV2 virus, but not dengue virus-2-derived non-structural protein 1 induced heparanase release from platelets. Conclusion Taken together, our findings suggest that the increase of heparanase activity in dengue patients is associated with endothelial glycocalyx degradation and plasma leakage. Furthermore, thrombin or DENV2 activated platelets may be considered as a potential source of heparanase.
Collapse
Affiliation(s)
- Baranca Buijsers
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Fadel Muhammad Garishah
- Department of Internal Medicine and the Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Center for Tropical and Infectious Diseases (CENTRID), Faculty of Medicine, Diponegoro University, Dr. Kariadi Hospital, Semarang, Indonesia
| | - Silvita Fitri Riswari
- Department of Internal Medicine and the Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Research Center for Care and Control of Infectious Disease (RC3ID), Universitas Padjadjaran, Bandung, Indonesia
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Rosalie M. van Ast
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Setyo Gundi Pramudo
- Department of Internal Medicine, Diponegoro National University Hospital, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
- Department of Internal Medicine, William Booth Hospital, Semarang, Indonesia
| | - Rahajeng N. Tunjungputri
- Department of Internal Medicine and the Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Center for Tropical and Infectious Diseases (CENTRID), Faculty of Medicine, Diponegoro University, Dr. Kariadi Hospital, Semarang, Indonesia
| | - Gijs J. Overheul
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ronald P. van Rij
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - André van der Ven
- Department of Internal Medicine and the Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Bachti Alisjahbana
- Research Center for Care and Control of Infectious Disease (RC3ID), Universitas Padjadjaran, Bandung, Indonesia
- Department of Internal Medicine, Hasan Sadikin General Hospital, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Muhammad Hussein Gasem
- Center for Tropical and Infectious Diseases (CENTRID), Faculty of Medicine, Diponegoro University, Dr. Kariadi Hospital, Semarang, Indonesia
- Department of Internal Medicine, Diponegoro National University Hospital, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
| | - Quirijn de Mast
- Department of Internal Medicine and the Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Johan van der Vlag
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- *Correspondence: Johan van der Vlag,
| |
Collapse
|
22
|
Effects of Hyperhomocysteinemia on the Platelet-Driven Contraction of Blood Clots. Metabolites 2021; 11:metabo11060354. [PMID: 34205914 PMCID: PMC8228611 DOI: 10.3390/metabo11060354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/21/2021] [Accepted: 05/30/2021] [Indexed: 12/17/2022] Open
Abstract
Hyperhomocysteinemia (HHcy) is associated with thrombosis, but the mechanistic links between them are not understood. We studied effects of homocysteine (Hcy) on clot contraction in vitro and in a rat model of HHcy. Incubation of blood with exogenous Hcy for 1 min enhanced clot contraction, while 15-min incubation led to a dose-dependent suppression of contraction. These effects were likely due to direct Hcy-induced platelet activation followed by exhaustion, as revealed by an increase in fibrinogen-binding capacity and P-selectin expression determined by flow cytometry. In the blood of rats with HHcy, clot contraction was enhanced at moderately elevated Hcy levels (10–50 μM), while at higher Hcy levels (>50 μM), the onset of clot contraction was delayed. HHcy was associated with thrombocytosis combined with a reduced erythrocyte count and hypofibrinogenemia. These data suggest that in HHcy, platelets get activated directly and indirectly, leading to enhanced clot contraction that is facilitated by the reduced content and resilience of fibrin and erythrocytes in the clot. The excessive platelet activation can lead to exhaustion and impaired contractility, which makes clots larger and more obstructive. In conclusion, HHcy modulates blood clot contraction, which may comprise an underappreciated pro- or antithrombotic mechanism.
Collapse
|
23
|
Pretorius E. Platelets in HIV: A Guardian of Host Defence or Transient Reservoir of the Virus? Front Immunol 2021; 12:649465. [PMID: 33968041 PMCID: PMC8102774 DOI: 10.3389/fimmu.2021.649465] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/06/2021] [Indexed: 01/28/2023] Open
Abstract
The immune and inflammatory responses of platelets to human immunodeficiency virus 1 (HIV-1) and its envelope proteins are of great significance to both the treatment of the infection, and to the comorbidities related to systemic inflammation. Platelets can interact with the HIV-1 virus itself, or with viral membrane proteins, or with dysregulated inflammatory molecules in circulation, ensuing from HIV-1 infection. Platelets can facilitate the inhibition of HIV-1 infection via endogenously-produced inhibitors of HIV-1 replication, or the virus can temporarily hide from the immune system inside platelets, whereby platelets act as HIV-1 reservoirs. Platelets are therefore both guardians of the host defence system, and transient reservoirs of the virus. Such reservoirs may be of particular significance during combination antiretroviral therapy (cART) interruption, as it may drive viral persistence, and result in significant implications for treatment. Both HIV-1 envelope proteins and circulating inflammatory molecules can also initiate platelet complex formation with immune cells and erythrocytes. Complex formation cause platelet hypercoagulation and may lead to an increased thrombotic risk. Ultimately, HIV-1 infection can initiate platelet depletion and thrombocytopenia. Because of their relatively short lifespan, platelets are important signalling entities, and could be targeted more directly during HIV-1 infection and cART.
Collapse
Affiliation(s)
- Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
24
|
Weisel JW, Litvinov RI. Visualizing thrombosis to improve thrombus resolution. Res Pract Thromb Haemost 2021; 5:38-50. [PMID: 33537528 PMCID: PMC7845077 DOI: 10.1002/rth2.12469] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/05/2020] [Accepted: 11/15/2020] [Indexed: 12/12/2022] Open
Abstract
The severity, course, and outcomes of thrombosis are determined mainly by the size and location of the thrombus, but studying thrombus structure and composition has been an important but challenging task. The substantial progress in determination of thrombus morphology has become possible due to new intravital imaging methodologies in combination with mechanical thrombectomy, which allows extraction of a fresh thrombus from a patient followed by microscopy. Thrombi have been found to contain various structural forms of fibrin along with platelet aggregates, leukocytes, and red blood cells, many of which acquire a polyhedral shape (polyhedrocytes) as a result of intravital platelet-driven contraction. The relative volume fractions of thrombus components and their structural forms vary substantially, depending on the clinical and pathogenic characteristics. This review summarizes recent research that describes quantitative and qualitative morphologic characteristics of arterial and venous thrombi that are relevant for the pathogenesis, prophylaxis, diagnosis, and treatment of thrombosis.
Collapse
Affiliation(s)
- John W. Weisel
- Department of Cell and Developmental BiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| | - Rustem I. Litvinov
- Department of Cell and Developmental BiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPAUSA
| |
Collapse
|
25
|
Impaired contraction of blood clots precedes and predicts postoperative venous thromboembolism. Sci Rep 2020; 10:18261. [PMID: 33106547 PMCID: PMC7589563 DOI: 10.1038/s41598-020-75234-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/13/2020] [Indexed: 12/18/2022] Open
Abstract
Deep vein thrombosis (DVT) is a common but unpredictable complication of surgical interventions. To reveal an association between the blood clot contraction (retraction) and the incidence of postoperative venous thrombosis, 78 patients with brain tumors that were operated on were studied, of which 23 (29%) were diagnosed with postoperative DVT. A clot contraction assay, along with other hemostatic and hematologic tests, was performed 1–3 days before the surgery and on the 1st day and 5–7th days after the surgery. On the 1st postoperative day, clot contraction was significantly suppressed in patients who subsequently developed DVT, compared to the patients without DVT. Importantly, this difference was observed at least 5 days before DVT had developed. The weakening of contraction on the 1st postoperative day was more pronounced in the DVT patients with malignant versus benign brain tumors, atherosclerosis, hypertension, as well as in patients receiving steroids before and during the operation. These results indicate that impaired clot contraction in the postoperative period is associated with imminent DVT, suggesting that it is a prothrombotic risk factor and promotional mechanism. The clot contraction assay has a predictive value in assessing the threat of postoperative thrombosis in patients with benign and malignant brain tumors.
Collapse
|
26
|
Mordakhanova ER, Nevzorova TA, Synbulatova GE, Rauova L, Weisel JW, Litvinov RI. Platelet Activation in Heparin-Induced Thrombocytopenia is Followed by Platelet Death via Complex Apoptotic and Non-Apoptotic Pathways. Int J Mol Sci 2020; 21:ijms21072556. [PMID: 32272655 PMCID: PMC7177543 DOI: 10.3390/ijms21072556] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/04/2020] [Accepted: 04/05/2020] [Indexed: 11/16/2022] Open
Abstract
Heparin-induced thrombocytopenia (HIT) is an adverse drug reaction characterized by thrombocytopenia and a high risk for venous or arterial thrombosis. HIT is caused by antibodies that recognize complexes of platelet factor 4 and heparin. The pathogenic mechanisms of this condition are not fully understood. In this study, we used flow cytometry, fluorimetry, and Western blot analysis to study the direct effects of pathogenic immune complexes containing platelet factor 4 on human platelets isolated by gel-filtration. HIT-like pathogenic immune complexes initially caused pronounced activation of platelets detected by an increased expression of phosphatidylserine and P-selectin. This activation was mediated either directly through the FcγRIIA receptors or indirectly via protease-activated receptor 1 (PAR1) receptors due to thrombin generated on or near the surface of activated platelets. The immune activation was later followed by the biochemical signs of cell death, such as mitochondrial membrane depolarization, up-regulation of Bax, down-regulation of Bcl-XL, and moderate activation of procaspase 3 and increased calpain activity. The results show that platelet activation under the action of HIT-like immune complexes is accompanied by their death through complex apoptotic and calpain-dependent non-apoptotic pathways that may underlie the low platelet count in HIT.
Collapse
Affiliation(s)
- Elmira R. Mordakhanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia; (E.R.M.); (T.A.N.); (G.E.S.)
| | - Tatiana A. Nevzorova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia; (E.R.M.); (T.A.N.); (G.E.S.)
| | - Gulnaz E. Synbulatova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia; (E.R.M.); (T.A.N.); (G.E.S.)
| | - Lubica Rauova
- The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
- Departments of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA;
| | - John W. Weisel
- Departments of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA;
| | - Rustem I. Litvinov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420008, Russia; (E.R.M.); (T.A.N.); (G.E.S.)
- Departments of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
- Correspondence:
| |
Collapse
|
27
|
Chernysh IN, Nagaswami C, Kosolapova S, Peshkova AD, Cuker A, Cines DB, Cambor CL, Litvinov RI, Weisel JW. The distinctive structure and composition of arterial and venous thrombi and pulmonary emboli. Sci Rep 2020; 10:5112. [PMID: 32198356 PMCID: PMC7083848 DOI: 10.1038/s41598-020-59526-x] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/21/2020] [Indexed: 11/09/2022] Open
Abstract
Although arterial and venous thromboembolic disorders are among the most frequent causes of mortality and morbidity, there has been little description of how the composition of thrombi and emboli depends on their vascular origin and age. We quantified the structure and composition of arterial and venous thrombi and pulmonary emboli using high-resolution scanning electron microscopy. Arterial thrombi contained a surprisingly large amount of fibrin, in addition to platelets. The composition of pulmonary emboli mirrored the most distal part of venous thrombi from which they originated, which differed from the structure of the body and head of the same thrombi. All thrombi and emboli contained few biconcave red blood cells but many polyhedrocytes or related forms of compressed red blood cells, demonstrating that these structures are a signature of clot contraction in vivo. Polyhedrocytes and intermediate forms comprised the major constituents of venous thrombi and pulmonary emboli. The structures within all of the thrombi and emboli were very tightly packed, in contrast to clots formed in vitro. There are distinctive, reproducible differences among arterial and venous thrombi and emboli related to their origin, destination and duration, which may have clinical implications for the understanding and treatment of thrombotic disorders.
Collapse
Affiliation(s)
- Irina N Chernysh
- University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | - Sofia Kosolapova
- University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | - Adam Cuker
- University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Douglas B Cines
- University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Carolyn L Cambor
- University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Rustem I Litvinov
- University of Pennsylvania School of Medicine, Philadelphia, PA, USA.,Kazan Federal University, Kazan, Russian Federation
| | - John W Weisel
- University of Pennsylvania School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
28
|
Stolla M, Bailey SL, Fang L, Fitzpatrick L, Gettinger I, Pellham E, Christoffel T. Effects of storage time prolongation on in vivo and in vitro characteristics of 4°C-stored platelets. Transfusion 2020; 60:613-621. [PMID: 32017135 DOI: 10.1111/trf.15669] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/30/2019] [Accepted: 12/09/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Cold (4°C)-stored platelets are currently under investigation for transfusion in bleeding patients. It is currently unknown how long cold-stored platelets can be stored for clinical applications. STUDY DESIGN AND METHODS Twenty three subjects were recruited. Twenty-one subjects were available for in vivo assessment and received indium-111 radiolabeled, cold-stored platelets. We investigated 5- (n = 5), 10- (n = 6), 15- (n = 5), and 20-day-stored (n = 5) platelets and obtained samples for in vitro testing at baseline and after the designated storage time. Twenty three units were available for in vitro testing. Five- and 7-day (n = 5 each), room temperature (RT)-stored platelets served as the current clinical standard control. RESULTS In vivo, we found a continuous decline in platelet recovery from 5 to 20 days. Platelet survival reached a low nadir after 10 days of storage. Ex vivo, we observed the maximum platelet αIIbβ3 integrin response to collagen at 5 days of cold storage, and we saw a continuous decline thereafter. However, platelet integrin activation and mitochondrial membrane integrity were better preserved after 20 days at 4°C, compared to 5 days at RT. Platelet metabolic parameters suggest comparable results between 20-day cold-stored platelets and 5- or 7-day RT-stored platelets. CONCLUSION In summary, we performed the first studies with extended, cold-stored, apheresis platelets in plasma for up to 20 days with a fresh comparator. Storing cold-stored platelets up to 20 days yields better results in vitro, but further studies in actively bleeding patients are needed to determine the best compromise between hemostatic efficacy and storage prolongation.
Collapse
Affiliation(s)
- Moritz Stolla
- Platelet Transfusion Research Laboratory, Bloodworks Northwest Research Institute, Seattle, Washington.,Department of Medicine, Division of Hematology, University of Washington School of Medicine, Seattle, Washington
| | - S Lawrence Bailey
- Platelet Transfusion Research Laboratory, Bloodworks Northwest Research Institute, Seattle, Washington
| | - Lydia Fang
- Platelet Transfusion Research Laboratory, Bloodworks Northwest Research Institute, Seattle, Washington
| | - Lynda Fitzpatrick
- Platelet Transfusion Research Laboratory, Bloodworks Northwest Research Institute, Seattle, Washington
| | - Irena Gettinger
- Platelet Transfusion Research Laboratory, Bloodworks Northwest Research Institute, Seattle, Washington
| | - Esther Pellham
- Platelet Transfusion Research Laboratory, Bloodworks Northwest Research Institute, Seattle, Washington
| | - Todd Christoffel
- Platelet Transfusion Research Laboratory, Bloodworks Northwest Research Institute, Seattle, Washington
| |
Collapse
|
29
|
Olie RH, van der Meijden PEJ, Spronk HMH, Ten Cate H. Antithrombotic Therapy: Prevention and Treatment of Atherosclerosis and Atherothrombosis. Handb Exp Pharmacol 2020; 270:103-130. [PMID: 32776281 DOI: 10.1007/164_2020_357] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Atherosclerosis is a multifactorial vascular disease that develops in the course of a lifetime. Numerous risk factors for atherosclerosis have been identified, mostly inflicting pro-inflammatory effects. Vessel injury, such as occurring during erosion or rupture of atherosclerotic lesions triggers blood coagulation, in attempt to maintain hemostasis (protect against bleeding). However, thrombo-inflammatory mechanisms may drive blood coagulation such that thrombosis develops, the key process underlying myocardial infarction and ischemic stroke (not due to embolization from the heart). In the blood coagulation system, platelets and coagulation proteins are both essential elements. Hyperreactivity of blood coagulation aggravates atherosclerosis in preclinical models. Pharmacologic inhibition of blood coagulation, either with platelet inhibitors, or better documented with anticoagulants, or both, limits the risk of thrombosis and may potentially reverse atherosclerosis burden, although the latter evidence is still based on animal experimentation.Patients at risk of atherothrombotic complications should receive a single antiplatelet agent (acetylsalicylic acid, ASA, or clopidogrel); those who survived an atherothrombotic event will be prescribed temporary dual antiplatelet therapy (ASA plus a P2Y12 inhibitor) in case of myocardial infarction (6-12 months), or stroke (<6 weeks), followed by a single antiplatelet agent indefinitely. High risk for thrombosis patients (such as those with peripheral artery disease) benefit from a combination of an anticoagulant and ASA. The price of gained efficacy is always increased risk of (major) bleeding; while tailoring therapy to individual needs may limit the risks to some extent, new generations of agents that target less critical elements of hemostasis and coagulation mechanisms are needed to maintain efficacy while reducing bleeding risks.
Collapse
Affiliation(s)
- R H Olie
- Internal Medicine and CARIM School for Cardiovascular Research, Maastricht University Medical Center, Maastricht, The Netherlands.,Thrombosis Expertise Center, Heart+ Cardiovascular Center, and Department of Biochemistry, Maastricht University Medical Center, Maastricht, The Netherlands
| | - P E J van der Meijden
- Thrombosis Expertise Center, Heart+ Cardiovascular Center, and Department of Biochemistry, Maastricht University Medical Center, Maastricht, The Netherlands
| | - H M H Spronk
- Thrombosis Expertise Center, Heart+ Cardiovascular Center, and Department of Biochemistry, Maastricht University Medical Center, Maastricht, The Netherlands
| | - H Ten Cate
- Internal Medicine and CARIM School for Cardiovascular Research, Maastricht University Medical Center, Maastricht, The Netherlands. .,Thrombosis Expertise Center, Heart+ Cardiovascular Center, and Department of Biochemistry, Maastricht University Medical Center, Maastricht, The Netherlands.
| |
Collapse
|
30
|
Arce NA, Li R. The secret afterlife of platelets. Haematologica 2019; 104:1699-1701. [PMID: 31473608 DOI: 10.3324/haematol.2019.224170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Nicholas A Arce
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine.,Graduate Program of Molecular and Systems Pharmacology, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA, USA
| | - Renhao Li
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine
| |
Collapse
|
31
|
Pretorius E. Platelets as Potent Signaling Entities in Type 2 Diabetes Mellitus. Trends Endocrinol Metab 2019; 30:532-545. [PMID: 31196615 DOI: 10.1016/j.tem.2019.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 12/19/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a multifactorial disease with a dysregulated circulating inflammatory molecule tendency. T2DM is closely associated with systemic inflammation, endothelial dysfunction, cardiovascular risk, and increased clotting susceptibility. Platelets have fundamental roles in the development and propagation of inflammation and cardiovascular risk. They signal through membrane receptors, resulting in (hyper)activation and release of inflammatory molecules from platelet compartments. This review highlights how circulating inflammatory molecules, acting as platelet receptor ligands, interact with platelets, causing platelets to be potent drivers of systemic inflammation. We conclude by suggesting that focused platelet research in T2DM is an important avenue to pursue to identify novel therapeutic targets, and that platelets could be used as cellular activity sensors themselves.
Collapse
Affiliation(s)
- Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, 7602, South Africa.
| |
Collapse
|
32
|
Nevzorova TA, Mordakhanova ER, Daminova AG, Ponomareva AA, Andrianova IA, Le Minh G, Rauova L, Litvinov RI, Weisel JW. Platelet factor 4-containing immune complexes induce platelet activation followed by calpain-dependent platelet death. Cell Death Discov 2019; 5:106. [PMID: 31263574 PMCID: PMC6591288 DOI: 10.1038/s41420-019-0188-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/29/2019] [Accepted: 06/05/2019] [Indexed: 01/23/2023] Open
Abstract
Heparin-induced thrombocytopenia (HIT) is a complication of heparin therapy sometimes associated with thrombosis. The hallmark of HIT is antibodies to the heparin/platelet factor 4 (PF4) complex that cause thrombocytopenia and thrombosis through platelet activation. Despite the clinical importance, the molecular mechanisms and late consequences of immune platelet activation are not fully understood. Here, we studied immediate and delayed effects of the complexes formed by human PF4 and HIT-like monoclonal mouse anti-human-PF4/heparin IgG antibodies (named KKO) on isolated human platelets in vitro. Direct platelet-activating effect of the KKO/PF4 complexes was corroborated by the overexpression of phosphatidylserine (PS) and P-selectin on the platelet surface. The immune platelet activation was accompanied by a decrease of the mitochondrial transmembrane potential (ΔΨm), concurrent with a significant gradual reduction of the ATP content in platelets, indicating disruption of energy metabolism. A combination of PS expression and mitochondrial depolarization induced by the PF4-containing immune complexes observed in a substantial fraction of platelets was considered as a sign of ongoing platelet death, as opposed to a subpopulation of activated live platelets with PS on the plasma membrane but normal ΔΨm. Both activated and dying platelets treated with KKO/PF4 formed procoagulant extracellular microvesicles bearing PS on their surface. Scanning and transmission electron microscopy revealed dramatic morphological changes of KKO/PF4-treated platelets, including their fragmentation, another indicator of cell death. Most of the effects of KKO/PF4 were prevented by an anti-FcγRII monoclonal antibody IV.3. The adverse functional and structural changes in platelets induced by the KKO/PF4 complexes were associated with strong time-dependent activation of calpain, but only trace cleavage of caspase 3. The results indicate that the pathogenic PF4-containing HIT-like immune complexes induce direct prothrombotic platelet activation via FcγRIIA receptors followed by non-apoptotic calpain-dependent death of platelets, which can be an important mechanism of thrombocytopenia during HIT development.
Collapse
Affiliation(s)
- Tatiana A. Nevzorova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., Kazan, Russian Federation 420008 Russia
| | - Elmira R. Mordakhanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., Kazan, Russian Federation 420008 Russia
| | - Amina G. Daminova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., Kazan, Russian Federation 420008 Russia
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevsky str., Kazan, Russian Federation 420111 Russia
| | - Anastasia A. Ponomareva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., Kazan, Russian Federation 420008 Russia
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, 2/31 Lobachevsky str., Kazan, Russian Federation 420111 Russia
| | - Izabella A. Andrianova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., Kazan, Russian Federation 420008 Russia
| | - Giang Le Minh
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., Kazan, Russian Federation 420008 Russia
| | - Lubica Rauova
- Children’s Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104 USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, 3401 Civic Center Blvd, Philadelphia, PA 19104 USA
| | - Rustem I. Litvinov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., Kazan, Russian Federation 420008 Russia
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, 421 Curie Boulevard, Philadelphia, PA 19104 USA
| | - John W. Weisel
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya St., Kazan, Russian Federation 420008 Russia
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, 421 Curie Boulevard, Philadelphia, PA 19104 USA
| |
Collapse
|