1
|
Shi Z, Ma R, Shan L, Tu H, Li Q, Su J, Lu F, Yu K, Geng Z, Slezak P, Zhou Z, Hu E, Shi S, Lan G, Xie R. Artificial Plateletoids Recruit Blood Proteins to Shield Symbiotic Thrombin: a Silk Fibroin/Calcium Interface Medicated Thrombin Generation and Preservation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2406909. [PMID: 39638929 DOI: 10.1002/smll.202406909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Breaking the constraints of thrombin during storage and in vivo applications remains challenging because of its low stability and sensitivity to environmental temperature and acidity. Herein, an artificial plateletoid is developed for in situ thrombin generation through a co-incubation approach with plasma in vitro, utilizing a silk fibroin/Ca2+ interface, to enhance the activity and stability of the generated thrombin. Notably, the enzymatic activity of the plateletoid thrombin platform is as high as 30 U g-1, leading to rapid clotting within 55 s, and it persisted for at least 90 days at as high as 37 °C. This considerably lessens the difficulties associated with maintaining the cold chain while storing and shipping thrombin formulations. Additionally, a gastric bleeding model confirmed that the plateletoid platform improved the acid resistance of thrombin by upregulating the pH of the gastric environment (pH 0.8), facilitating oral delivery of thrombin for effective hemorrhage control in highly acidic stomach conditions. This pioneering study addresses the constraints of thrombin in storage and in vivo applications and may provide a basis for further research on biological storage and delivery approaches.
Collapse
Affiliation(s)
- Zhenghui Shi
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Ruiyao Ma
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Lianqi Shan
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Hongyu Tu
- Chongqing Customs Technology Center, Chongqing, 400044, China
| | - Qing Li
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Jing Su
- College of Textile Science and Engineering, Jiangnan University, Wuxi, 214122, China
| | - Fei Lu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Kun Yu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Zhen Geng
- Institute of Translational Medicine, Organoid Research Center, National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Paul Slezak
- Ludwig Boltzmann Institute for Traumatology, AUVA Research Center, Vienna, 1200, Austria
| | - Zhihang Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Enling Hu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
- School of Fashion and Textiles, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Shuo Shi
- School of Fashion and Textiles, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Guangqian Lan
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Ruiqi Xie
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
- Ludwig Boltzmann Institute for Traumatology, AUVA Research Center, Vienna, 1200, Austria
| |
Collapse
|
2
|
Greenman JH, Moss L, Chakraborty S, Whitehead BJ, Palmfedt J, Nejsum P, Hewitson JP, Hitchcock IS. Brief report: Chronic murine schistosomiasis causes aberrant hemostasis. Exp Hematol 2024; 142:104689. [PMID: 39615580 DOI: 10.1016/j.exphem.2024.104689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/19/2024]
Abstract
Schistosomiasis afflicts >250 million people worldwide, leading to an annual loss of >3 million disability-adjusted life years. Schistosoma mansoni causes intestinal schistosomiasis with parasite eggs either transversing intestinal tissue or lodging within the liver and other organs, causing intestinal hemorrhage and liver pathology. Large (∼1 cm) adult worms survive for years within blood vessels, but we lack a clear understanding of their impact on hemostasis. We used a chronic mouse model of schistosomiasis to determine the impact on platelet numbers, phenotype and function. Hemostatic function was assessed by platelet phenotyping (flow cytometry and proteomics), whole blood aggregometry, and longitudinal coagulometry. Although platelets from schistosome-infected mice lack elevated surface P-selectin and activated αIIbβ3, unbiased proteomic analysis reveals infection-induced increases in MHC-I, IgM and IgG antibodies, and complement components. Whole blood from schistosome-infected mice spontaneously aggregates in the absence of exogenous agonists. Conversely, prothrombin and activated partial thromboplastin times are prolonged at the chronic stage of infection (10-12 weeks). A mouse model of S. mansoni infection shows wide-ranging changes in hemostatic function which may have clinically relevant implications for populations in endemic regions.
Collapse
Affiliation(s)
- Joanna H Greenman
- Department of Biology, University of York, Heslington, York, United Kingdom; York Biomedical Research Institute, University of York, Heslington, York, United Kingdom
| | - Lucie Moss
- Department of Biology, University of York, Heslington, York, United Kingdom; York Biomedical Research Institute, University of York, Heslington, York, United Kingdom
| | - Shinjini Chakraborty
- Department of Biology, University of York, Heslington, York, United Kingdom; York Biomedical Research Institute, University of York, Heslington, York, United Kingdom
| | - Bradley J Whitehead
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Johan Palmfedt
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Peter Nejsum
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - James P Hewitson
- Department of Biology, University of York, Heslington, York, United Kingdom; York Biomedical Research Institute, University of York, Heslington, York, United Kingdom.
| | - Ian S Hitchcock
- Department of Biology, University of York, Heslington, York, United Kingdom; York Biomedical Research Institute, University of York, Heslington, York, United Kingdom.
| |
Collapse
|
3
|
Wurtzel JGT, Gray BD, Pak KY, Zhao X, Ma P, McKenzie SE, Tanujaya M, Rizzo V, Del Carpio-Cano F, Rao AK, Lee-Gau Chong P, Goldfinger LE. Phosphatidylserine-blocking nanoparticles inhibit thrombosis without increased bleeding in mice. J Thromb Haemost 2024:S1538-7836(24)00613-5. [PMID: 39423958 DOI: 10.1016/j.jtha.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/24/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Phosphatidylserine (PS) is a procoagulant phospholipid enriched on surfaces of activated vascular cells including platelets, endothelium, monocytes, and microvesicles. As a molecular driver of thrombosis accessible to drug blockade, PS is an attractive pharmacologic target for modulating thrombogenesis, with potentially reduced bleeding risk compared to anticoagulant and antiplatelet therapies. OBJECTIVES Test antithrombotic capabilities of a liposomal formulation, Zn-dipicolylamine cyanine-3[22,22]/1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (molar ratio, 3:97), designated as DPAL, which we previously described binds selectively to PS-enriched cell surfaces, compared with effects on bleeding, in mouse models. METHODS PS-dependent DPAL binding to human and murine platelets was tested in vitro. Thrombosis and bleeding after DPAL intravenous administration were tested in C57Bl/6J mice following FeCl3 carotid arterial injury and tail tip amputation, respectively. Incorporation in hemostatic clots was investigated in the cremaster muscle laser injury model. Toxicity was tested by direct exposure to human endothelial cell cultures. RESULTS DPAL bound agonist-stimulated, PS-positive human and murine platelets, blocked by Annexin V or Ano6 deletion, which ablate PS exposure. DPAL prolonged prothrombin time, but did not prevent thrombin-induced fibrinogen receptor activation or aggregation, nor alter blood cell counts including platelets. Following arteriolar laser injury, DPAL bound wound surfaces and edges without destabilizing plugs. DPAL dose-dependently blocked FeCl3-induced arterial thrombosis but did not substantially increase bleeding, or induce endothelial cell death. CONCLUSION DPAL reduces thrombogenesis with minimal effects on bleeding in mouse models via selective binding to PS. DPAL may support novel approaches to modulate pathogenic thrombin generation with improved safety profiles in multiple contexts.
Collapse
Affiliation(s)
- Jeremy G T Wurtzel
- Cardeza Foundation for Hematologic Research, Department of Medicine, Division of Hematology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Brian D Gray
- Molecular Targeting Technologies Inc, West Chester, Pennsylvania, USA
| | - Koon Y Pak
- Molecular Targeting Technologies Inc, West Chester, Pennsylvania, USA
| | - Xuefei Zhao
- Cardeza Foundation for Hematologic Research, Department of Medicine, Division of Hematology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Peisong Ma
- Cardeza Foundation for Hematologic Research, Department of Medicine, Division of Hematology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Steven E McKenzie
- Cardeza Foundation for Hematologic Research, Department of Medicine, Division of Hematology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Michelle Tanujaya
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Fabiola Del Carpio-Cano
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - A Koneti Rao
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA; Department of Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Parkson Lee-Gau Chong
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Lawrence E Goldfinger
- Cardeza Foundation for Hematologic Research, Department of Medicine, Division of Hematology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
4
|
Knauss EA, Guci J, Luc N, Disharoon D, Huang GH, Gupta AS, Nieman MT. Mice with Reduced PAR4 Reactivity show Decreased Venous Thrombosis and Platelet Procoagulant Activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.617127. [PMID: 39463946 PMCID: PMC11507748 DOI: 10.1101/2024.10.14.617127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Background Hypercoagulation and thrombin generation are major risk factors for venous thrombosis. Sustained thrombin signaling through PAR4 promotes platelet activation, phosphatidylserine exposure, and subsequent thrombin generation. A single-nucleotide polymorphism in PAR4 (rs2227376) changes proline to leucine extracellular loop 3 (P310L), which decreases PAR4 reactivity and is associated with a lower risk for venous thromboembolism (VTE) in a GWAS meta-analysis. Objective The goal of this study is to determine the mechanism for the association of rs2227376 with reduced risk for VTE in using mice with a homologous mutation (PAR4-P322L). Methods Venous thrombosis was examined using our recently generated PAR4-P322L mice in the inferior vena cava stasis and stenosis models. Coagulation and clot stability was measured using rotational thromboelastometry (ROTEM). Thrombin generating potential was measured in platelet-rich plasma. Phosphatidylserine surface expression and platelet-neutrophil aggregates were analyzed using flow cytometry. Results PAR4P/L and PAR4L/L had reduced incidence and size of venous clots at 48 hours. PAR4P/L and PAR4L/L platelets had progressively decreased phosphatidylserine in response to thrombin and convulxin, which led to decreased thrombin generation and decreased PAR4-mediated platelet-neutrophil aggregation. Conclusions The leucine allele in extracellular loop 3, PAR4-322L leads to fewer procoagulant platelets and decreased endogenous thrombin potential. This decreased ability to generate thrombin offers a mechanism for PAR4's role in VTE highlighting a key role for PAR4 signaling.
Collapse
Affiliation(s)
- Elizabeth A. Knauss
- Case Western Reserve University, School of Medicine, Department of Pharmacology, Cleveland, OH United States
| | - Johana Guci
- Case Western Reserve University, School of Medicine, Department of Pharmacology, Cleveland, OH United States
| | - Norman Luc
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, OH United States
| | - Dante Disharoon
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, OH United States
| | - Grace H. Huang
- Case Western Reserve University, School of Medicine, Department of Pharmacology, Cleveland, OH United States
| | - Anirban Sen Gupta
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, OH United States
| | - Marvin T. Nieman
- Case Western Reserve University, School of Medicine, Department of Pharmacology, Cleveland, OH United States
| |
Collapse
|
5
|
Lira AL, Kohs TC, Moellmer SA, Shatzel JJ, McCarty OJ, Puy C. Substrates, Cofactors, and Cellular Targets of Coagulation Factor XIa. Semin Thromb Hemost 2024; 50:962-969. [PMID: 36940715 PMCID: PMC11069399 DOI: 10.1055/s-0043-1764469] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2023]
Abstract
Coagulation factor XI (FXI) has increasingly been shown to play an integral role in several physiologic and pathological processes. FXI is among several zymogens within the blood coagulation cascade that are activated by proteolytic cleavage, with FXI converting to the active serine protease form (FXIa). The evolutionary origins of FXI trace back to duplication of the gene that transcribes plasma prekallikrein, a key factor in the plasma kallikrein-kinin system, before further genetic divergence led to FXI playing a unique role in blood coagulation. While FXIa is canonically known for activating the intrinsic pathway of coagulation by catalyzing the conversion of FIX into FIXa, it is promiscuous in nature and has been shown to contribute to thrombin generation independent of FIX. In addition to its role in the intrinsic pathway of coagulation, FXI also interacts with platelets, endothelial cells, and mediates the inflammatory response through activation of FXII and cleavage of high-molecular-weight kininogen to generate bradykinin. In this manuscript, we critically review the current body of knowledge surrounding how FXI navigates the interplay of hemostasis, inflammatory processes, and the immune response and highlight future avenues for research. As FXI continues to be clinically explored as a druggable therapeutic target, understanding how this coagulation factor fits into physiological and disease mechanisms becomes increasingly important.
Collapse
Affiliation(s)
- André L. Lira
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Tia C.L. Kohs
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Samantha A. Moellmer
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Joseph J. Shatzel
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Divison of Hematology and Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Owen J.T. McCarty
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Divison of Hematology and Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Cristina Puy
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Divison of Hematology and Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
6
|
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
|
7
|
Ballard-Kordeliski A, Lee RH, O’Shaughnessy EC, Kim PY, Jones SR, Pawlinski R, Flick MJ, Paul DS, Mackman N, Adalsteinsson DA, Bergmeier W. 4D intravital imaging studies identify platelets as the predominant cellular procoagulant surface in a mouse hemostasis model. Blood 2024; 144:1116-1126. [PMID: 38820498 PMCID: PMC11406176 DOI: 10.1182/blood.2023022608] [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: 09/26/2023] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 06/02/2024] Open
Abstract
ABSTRACT Interplay between platelets, coagulation factors, endothelial cells (ECs), and fibrinolytic factors is necessary for effective hemostatic plug formation. This study describes a 4-dimensional (4D) imaging platform to visualize and quantify hemostatic plug components in mice with high spatiotemporal resolution. Fibrin accumulation after laser-induced vascular injury was observed at the platelet plug-EC interface, controlled by the antagonistic balance between fibrin generation and breakdown. We observed less fibrin accumulation in mice expressing low levels of tissue factor or F12-/-mice compared with controls, whereas increased fibrin accumulation, including on the vasculature adjacent to the platelet plug, was observed in plasminogen-deficient mice or wild-type mice treated with tranexamic acid. Phosphatidylserine (PS), a membrane lipid critical for the assembly of coagulation factors, was first detected at the platelet plug-EC interface, followed by exposure across the endothelium. Impaired PS exposure resulted in a significant reduction in fibrin accumulation in cyclophilin D-/-mice. Adoptive transfer studies demonstrated a key role for PS exposure on platelets, and to a lesser degree on ECs, in fibrin accumulation during hemostatic plug formation. Together, these studies suggest that (1) platelets are the functionally dominant procoagulant cellular surface, and (2) plasmin is critical for limiting fibrin accumulation at the site of a forming hemostatic plug.
Collapse
Affiliation(s)
- Abigail Ballard-Kordeliski
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Robert H. Lee
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Ellen C. O’Shaughnessy
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Paul Y. Kim
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, Canada
| | - Summer R. Jones
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Rafal Pawlinski
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Matthew J. Flick
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - David S. Paul
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Nigel Mackman
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - David A. Adalsteinsson
- Department of Mathematics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
8
|
Puspitasari YM, Ministrini S, Han J, Karch C, Prisco F, Liberale L, Bengs S, Akhmedov A, Montecucco F, Beer JH, Lüscher TF, Bongiovanni D, Camici GG. Hutchinson-Gilford progeria syndrome mice display accelerated arterial thrombus formation and increased platelet reactivity. Thromb Res 2024; 241:109100. [PMID: 39032390 DOI: 10.1016/j.thromres.2024.109100] [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: 12/18/2023] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
INTRODUCTION Hutchinson-Gilford Progeria Syndrome (HGPS) is an ultra-rare premature aging genetic disorder caused by a point mutation in the lamin A gene, LMNA. Children with HGPS display short lifespans and typically die due to myocardial infarction or ischemic stroke, both acute cardiovascular events that are tightly linked to arterial thrombosis. Despite this fact, the effect of the classic HGPS LMNA gene mutation on arterial thrombosis remains unknown. METHODS Heterozygous LmnaG609G knock-in (LmnaG609G/+) mice, yielding an equivalent classic mutation observed in HGPS patients (c.1824C>T; pG608G mutation in the human LMNA gene) and corresponding wild-type (WT) control littermates underwent photochemically laser-induced carotid injury to trigger thrombosis. Coagulation and fibrinolytic factors were measured. Furthermore, platelet activation and reactivity were investigated. RESULTS LmnaG609G/+ mice displayed accelerated arterial thrombus formation, as underlined by shortened time to occlusion compared to WT littermates. Levels of factors involved in the coagulation and fibrinolytic system were comparable between groups, while LmnaG609G/+ animals showed higher plasma levels of thrombin-antithrombin complex and lower levels of antithrombin. Bone marrow analysis showed larger megakaryocytes in progeric mice. Lastly, enhanced platelet activation upon adenosine diphosphate, collagen-related peptide, and thrombin stimulation was observed in LmnaG609G/+ animals compared to the WT group, indicating a higher platelet reactivity in progeric animals. CONCLUSIONS LMNA mutation in HGPS mice accelerates arterial thrombus formation, which is mediated, at least in part, by enhanced platelet reactivity, which consequently augments thrombin generation. Given the wide spectrum of antiplatelet agents available clinically, further investigation is warranted to consider the most suitable antiplatelet regimen for children with HGPS to mitigate disease mortality and morbidity.
Collapse
Affiliation(s)
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Internal Medicine, Angiology and Atherosclerosis, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Jiaying Han
- Department of Internal Medicine I, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Caroline Karch
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Francesco Prisco
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Switzerland
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Susan Bengs
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital of Baden, Baden, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Cardiology, Royal Brompton & Harefield Hospitals, National Heart & Lung Institute, Imperial College, London, United Kingdom
| | - Dario Bongiovanni
- Department of Internal Medicine I, Cardiology, University Hospital Augsburg, University of Augsburg, Augsburg, Germany; Department of Cardiovascular Medicine, Humanitas Clinical and Research Center IRCCS and Humanitas University, Rozzano, Milan, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Research and Education, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
9
|
Yadav P, Beura SK, Panigrahi AR, Kulkarni PP, Yadav MK, Munshi A, Singh SK. Lysophosphatidylcholine induces oxidative stress and calcium-mediated cell death in human blood platelets. Cell Biol Int 2024; 48:1266-1284. [PMID: 38837523 DOI: 10.1002/cbin.12192] [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: 12/15/2023] [Revised: 05/06/2024] [Accepted: 05/11/2024] [Indexed: 06/07/2024]
Abstract
Platelets are essential component of circulation that plays a major role in hemostasis and thrombosis. During activation and its demise, platelets release platelet-derived microvesicles, with lysophosphatidylcholine (LPC) being a prominent component in their lipid composition. LPC, an oxidized low-density lipoprotein, is involved in cellular metabolism, but its higher level is implicated in pathologies like atherosclerosis, diabetes, and inflammatory disorders. Despite this, its impact on platelet function remains relatively unexplored. To address this, we studied LPC's effects on washed human platelets. A multimode plate reader was employed to measure reactive oxygen species and intracellular calcium using H2DCF-DA and Fluo-4-AM, respectively. Flow cytometry was utilized to measure phosphatidylserine expression, mitochondrial membrane potential (ΔΨm), and mitochondrial permeability transition pore (mPTP) formation using FITC-Annexin V, JC-1, and CoCl2/calcein-AM, respectively. Additionally, platelet morphology and its ultrastructure were observed via phase contrast and electron microscopy. Sonoclot and light transmission aggregometry were employed to examine fibrin formation and platelet aggregation, respectively. The findings demonstrate that LPC induced oxidative stress and increased intracellular calcium in platelets, resulting in increased phosphatidylserine expression and reduced ΔΨm. LPC triggered caspase-independent platelet death and mPTP opening via cytosolic and mitochondrial calcium, along with microvesiculation and reduced platelet counts. LPC increased the platelet's size, adopting a balloon-shaped morphology, causing membrane fragmentation and releasing its cellular contents, while inducing a pro-coagulant phenotype with increased fibrin formation and reduced integrin αIIbβ3 activation. Conclusively, this study reveals LPC-induced oxidative stress and calcium-mediated platelet death, necrotic in nature with pro-coagulant properties, potentially impacting inflammation and repair mechanisms during vascular injury.
Collapse
Affiliation(s)
- Pooja Yadav
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
| | - Samir K Beura
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
| | - Abhishek R Panigrahi
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
| | - Paresh P Kulkarni
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mithlesh K Yadav
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Ghudda, Bathinda, India
| | - Sunil K Singh
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, India
| |
Collapse
|
10
|
Zhang Y, Zeng J, Bao S, Zhang B, Li X, Wang H, Cheng Y, Zhang H, Zu L, Xu X, Xu S, Song Z. Cancer progression and tumor hypercoagulability: a platelet perspective. J Thromb Thrombolysis 2024; 57:959-972. [PMID: 38760535 DOI: 10.1007/s11239-024-02993-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/26/2024] [Indexed: 05/19/2024]
Abstract
Venous thromboembolism, which is common in cancer patients and accompanies or even precedes malignant tumors, is known as cancer-related thrombosis and is an important cause of cancer- associated death. At present, the exact etiology of the elevated incidence of venous thrombosis in cancer patients remains elusive. Platelets play a crucial role in blood coagulation, which is intimately linked to the development of arterial thrombosis. Additionally, platelets contribute to tumor progression and facilitate immune evasion by tumors. Tumor cells can interact with the coagulation system through various mechanisms, such as producing hemostatic proteins, activating platelets, and directly adhering to normal cells. The relationship between platelets and malignant tumors is also significant. In this review article, we will explore these connections.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingtong Zeng
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Shihao Bao
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Bo Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xianjie Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Hanqing Wang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuan Cheng
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Hao Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Lingling Zu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaohong Xu
- Colleges of Nursing, Tianjin Medical University, Tianjin, China
| | - Song Xu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China.
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China.
| | - Zuoqing Song
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China.
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
11
|
Huang Y, Wang J, Guo Y, Shen L, Li Y. Fibrinogen binding to activated platelets and its biomimetic thrombus-targeted thrombolytic strategies. Int J Biol Macromol 2024; 274:133286. [PMID: 38908635 DOI: 10.1016/j.ijbiomac.2024.133286] [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: 11/24/2023] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
Thrombosis is associated with various fatal arteriovenous syndromes including ischemic stroke, myocardial infarction, and pulmonary embolism. However, current clinical thrombolytic treatment strategies still have many problems in targeting and safety to meet the thrombolytic therapy needs. Understanding the molecular mechanism that underlies thrombosis is critical in developing effective thrombolytic strategies. It is well known that platelets play a central role in thrombosis and the binding of fibrinogen to activated platelets is a common pathway in the process of clot formation. Based on this, a concept of biomimetic thrombus-targeted thrombolytic strategy inspired from fibrinogen binding to activated platelets in thrombosis was proposed, which could selectively bind to activated platelets at a thrombus site, thus enabling targeted delivery and local release of thrombolytic agents for effective thrombolysis. In this review, we first summarized the main characteristics of platelets and fibrinogen, and then introduced the classical molecular mechanisms of thrombosis, including platelet adhesion, platelet activation and platelet aggregation through the interactions of activated platelets with fibrinogen. In addition, we highlighted the recent advances in biomimetic thrombus-targeted thrombolytic strategies which inspired from fibrinogen binding to activated platelets in thrombosis. The possible future directions and perspectives in this emerging area are briefly discussed.
Collapse
Affiliation(s)
- Yu Huang
- Department of Radiology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, PR China.
| | - Jiahua Wang
- Department of Radiology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, PR China
| | - Yuanyuan Guo
- Department of Radiology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, PR China
| | - Lingyue Shen
- Department of Oral & Maxillofacial-Head & Neck Oncology, Department of Laser and Aesthetic Medicine, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stoma-tology & Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai 200011, PR China.
| | - Yuehua Li
- Department of Radiology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, PR China.
| |
Collapse
|
12
|
Price AD, Chae RC, Wallen TE, Becker ER, Baucom MR, Schuster RM, England L, Pritts TA, Goodman MD. Direct red blood cell effect on thrombosis is dependent on the interaction of tissue factor and calcium with membrane phosphatidylserine. J Trauma Acute Care Surg 2024; 97:57-64. [PMID: 38605437 PMCID: PMC11199102 DOI: 10.1097/ta.0000000000004340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
BACKGROUND Prior literature has implicated red blood cells (RBCs) in the initiation of thrombosis and suggests that posttransfusion hypercoagulability may occur secondary to the effects of RBCs. Elevated serum tissue factor is a known sequelae of acute trauma. Phosphatidylserine (PS) is a prothrombotic phospholipid present within the RBC cell membrane. We hypothesized that RBC aggregation is dependent on the interaction between RBC membrane bound (exposed) PS, extracellular calcium, and tissue factor. METHODS Human whole blood (WB) was separated into components, including RBCs and platelet-rich plasma (PRP). Whole blood, PRP, and RBCs underwent impedance aggregometry utilizing arachidonic acid (AA), ADP, collagen, calcium, and tissue factor (TF)-based agonists. Red blood cells then underwent impedance aggregometry utilizing combined calcium and TF agonists. Red blood cells were pretreated with Annexin V, a known PS blocking agent, and underwent impedance aggregometry with combined calcium and TF agonists to determine if the mechanism of calcium/TF-induced RBC aggregability is dependent on PS. Red blood cells treated with calcium, TF, calcium+TF, and pre-treated with Annexin V followed by calcium+TF were perfused through an in vitro model of pulmonary microcirculatory flow. RESULTS Red blood cell aggregation was significantly higher than that of WB and PRP when utilizing a TF agonist, an effect unique to TF. The combination of calcium and TF demonstrated significantly higher RBC aggregation than either agonist alone. Pretreatment with Annexin V resulted in a significantly reduced aggregability of RBC following treatment with TF + calcium. Red blood cells aged to 42 days did not exhibit significant change in aggregation. Exposure to calcium and TF significantly reduced time to thrombosis of RBCs perfused through a pulmonary microcirculatory model. CONCLUSION Treatment with both TF and calcium synergistically induces RBC aggregation. Phosphatidylserine appears to play an integral role in the TF/calcium-based, age-independent RBC aggregation response. Red blood cells treated with TF + calcium exhibit more rapid thrombus formation in an in vitro model of pulmonary microcirculatory perfusion.
Collapse
Affiliation(s)
- Adam D Price
- From the Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Hoy CK, NaveenKumar SK, Navaz SA, Sugur K, Yalavarthi S, Sarosh C, Smith T, Kmetova K, Chong E, Peters NF, Rysenga CE, Norman GL, Figueroa-Parra G, Nelson D, Girard J, Ahmed AZ, Schaefer JK, Gudjonsson JE, Kahlenberg JM, Madison JA, Knight JS, Crowson CS, Duarte-García A, Zuo Y. Calprotectin Impairs Platelet Survival in Patients With Primary Antiphospholipid Syndrome. Arthritis Rheumatol 2024; 76:928-935. [PMID: 38225923 PMCID: PMC11136595 DOI: 10.1002/art.42801] [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: 06/26/2023] [Revised: 11/29/2023] [Accepted: 01/12/2024] [Indexed: 01/17/2024]
Abstract
OBJECTIVE While thrombosis and pregnancy loss are the best-known clinical features of antiphospholipid syndrome (APS), many patients also exhibit "extra-criteria" manifestations, such as thrombocytopenia. The mechanisms that drive APS thrombocytopenia are not completely understood, and no clinical biomarkers are available for predicting antiphospholipid antibody (aPL)-mediated thrombocytopenia. Calprotectin is a heterodimer of S100A8 and S100A9 that is abundant in the neutrophil cytoplasm and released upon proinflammatory neutrophil activation. Here, we sought to evaluate the presence, clinical associations, and potential mechanistic roles of circulating calprotectin in a cohort of primary APS and aPL-positive patients. METHODS Levels of circulating calprotectin were determined in plasma by the QUANTA Flash chemiluminescent assay. A viability dye-based platelet assay was used to assess the potential impact of calprotectin on aPL-mediated thrombocytopenia. RESULTS Circulating calprotectin was measured in 112 patients with primary APS and 30 aPL-positive (without APS criteria manifestations or lupus) patients as compared to patients with lupus (without APS), patients with unprovoked venous thrombosis (without aPL), and healthy controls. Levels of calprotectin were higher in patients with primary APS and aPL-positive patients compared to healthy controls. After adjustment for age and sex, calprotectin level correlated positively with absolute neutrophil count (r = 0.41, P < 0.001), positively with C-reactive protein level (r = 0.34, P = 0.002), and negatively with platelet count (r = -0.24, P = 0.004). Mechanistically, we found that calprotectin provoked aPL-mediated thrombocytopenia by engaging platelet surface toll-like receptor 4 and activating the NLRP3-inflammasome, thereby reducing platelet viability in a caspase-1-dependent manner. CONCLUSION These data suggest that calprotectin has the potential to be a functional biomarker and a new therapeutic target for APS thrombocytopenia.
Collapse
Affiliation(s)
- Claire K. Hoy
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Sherwin A. Navaz
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kavya Sugur
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Srilakshmi Yalavarthi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Cyrus Sarosh
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Tristin Smith
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Katarina Kmetova
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Emily Chong
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Noah F. Peters
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christine E. Rysenga
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Gary L. Norman
- Headquarters & Technology Center Autoimmunity, Werfen, San Diego, CA, USA
| | - Gabriel Figueroa-Parra
- Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Dava Nelson
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jennifer Girard
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Asra Z. Ahmed
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jordan K. Schaefer
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - J. Michelle Kahlenberg
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jacqueline A. Madison
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason S. Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Cynthia S. Crowson
- Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Alí Duarte-García
- Division of Rheumatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Yu Zuo
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
14
|
Zhou J, Rico MC, Rauova L, Poncz M, Essex DW. Thioredoxin-related transmembrane protein 1 negatively regulates coagulation and phosphatidylserine exposure. Res Pract Thromb Haemost 2024; 8:102472. [PMID: 39036672 PMCID: PMC11260325 DOI: 10.1016/j.rpth.2024.102472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/02/2024] [Accepted: 06/06/2024] [Indexed: 07/23/2024] Open
Abstract
Background Five secreted platelet protein disulfide isomerases (PDIs) and 1 transmembrane PDI regulate platelet function and thrombosis. Thioredoxin-related transmembrane protein 1 (TMX1) was the first member of the PDI family found to negatively regulate platelet aggregation and platelet accumulation in vivo. The effect of TMX1 on coagulation is unknown. Objectives To determine the effect of TMX1 on coagulation. Methods TMX1-/- mice were used to study platelet accumulation and fibrin deposition in vivo in the laser-induced thrombosis injury model. Annexin V deposition at the site of vascular injury was studied using conditional TMX1 knockout mice. Annexin V binding to platelets was studied using human platelets, anti-TMX1 antibodies, and TMX1-deficient platelets. Results TMX1-/- mice had increased fibrin deposition that was reversed with infusion of recombinant TMX1. Infusion of recombinant TMX1 inhibited platelet accumulation and fibrin deposition in wild-type mice and inhibited fibrin deposition in β3-null mice. Platelet accumulation is absent in β3-null mice, suggesting that TMX1 inhibits coagulation independently of platelets. Annexin V binding was increased in activated human platelets incubated with an anti-TMX1 antibody and mouse platelets lacking TMX1. Addition of recombinant TMX1 decreased annexin V binding to platelets. Annexin V binding was increased at the site of vascular injury in Tie2-Cre/TMX1fl/fl mice deficient in endothelial cell TMX1. Conclusion TMX1 decreases coagulation at the site of vascular injury and negatively regulates phosphatidylserine exposure on endothelial cells and platelets.
Collapse
Affiliation(s)
- Junsong Zhou
- The Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu, China
- Division of Hematology, Department of Medicine, Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Mario C. Rico
- Division of Hematology, Department of Medicine, Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Lubica Rauova
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mortimer Poncz
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David W. Essex
- Division of Hematology, Department of Medicine, Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
15
|
Guerreiro EM, Kruglik SG, Swamy S, Latysheva N, Østerud B, Guigner JM, Sureau F, Bonneau S, Kuzmin AN, Prasad PN, Hansen JB, Hellesø OG, Snir O. Extracellular vesicles from activated platelets possess a phospholipid-rich biomolecular profile and enhance prothrombinase activity. J Thromb Haemost 2024; 22:1463-1474. [PMID: 38266680 DOI: 10.1016/j.jtha.2024.01.004] [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: 05/08/2023] [Revised: 12/12/2023] [Accepted: 01/11/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Extracellular vesicles (EVs), in particular those derived from activated platelets, are associated with a risk of future venous thromboembolism. OBJECTIVES To study the biomolecular profile and function characteristics of EVs from control (unstimulated) and activated platelets. METHODS Biomolecular profiling of single or very few (1-4) platelet-EVs (control/stimulated) was performed by Raman tweezers microspectroscopy. The effects of such EVs on the coagulation system were comprehensively studied. RESULTS Raman tweezers microspectroscopy of platelet-EVs followed by biomolecular component analysis revealed for the first time 3 subsets of EVs: (i) protein rich, (ii) protein/lipid rich, and (iii) lipid rich. EVs from control platelets presented a heterogeneous biomolecular profile, with protein-rich EVs being the main subset (58.7% ± 3.5%). Notably, the protein-rich subset may contain a minor contribution from other extracellular particles, including protein aggregates. In contrast, EVs from activated platelets were more homogeneous, dominated by the protein/lipid-rich subset (>85%), and enriched in phospholipids. Functionally, EVs from activated platelets increased thrombin generation by 52.4% and shortened plasma coagulation time by 34.6% ± 10.0% compared with 18.6% ± 13.9% mediated by EVs from control platelets (P = .015). The increased procoagulant activity was predominantly mediated by phosphatidylserine. Detailed investigation showed that EVs from activated platelets increased the activity of the prothrombinase complex (factor Va:FXa:FII) by more than 6-fold. CONCLUSION Our study reports a novel quantitative biomolecular characterization of platelet-EVs possessing a homogenous and phospholipid-enriched profile in response to platelet activation. Such characteristics are accompanied with an increased phosphatidylserine-dependent procoagulant activity. Further investigation of a possible role of platelet-EVs in the pathogenesis of venous thromboembolism is warranted.
Collapse
Affiliation(s)
- Eduarda M Guerreiro
- Thrombosis Research Group, Institute of Clinical Medicine, Univesitet i Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Sergei G Kruglik
- Laboratoire Jean Perrin, Institut de Biologie Paris-Seine, Sorbonne Université, Centre National de la Recherche Scientifique, Paris, France.
| | - Samantha Swamy
- Thrombosis Research Group, Institute of Clinical Medicine, Univesitet i Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Nadezhda Latysheva
- Thrombosis Research Group, Institute of Clinical Medicine, Univesitet i Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Bjarne Østerud
- Thrombosis Research Group, Institute of Clinical Medicine, Univesitet i Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Jean-Michel Guigner
- L'Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, Sorbonne Université, Centre National de la Recherche Scientifique, Institut de Recherche pour le Développement, Muséum National d'Histoire Naturelle, Paris, France
| | - Franck Sureau
- Laboratoire Jean Perrin, Institut de Biologie Paris-Seine, Sorbonne Université, Centre National de la Recherche Scientifique, Paris, France
| | - Stephanie Bonneau
- Laboratoire Jean Perrin, Institut de Biologie Paris-Seine, Sorbonne Université, Centre National de la Recherche Scientifique, Paris, France
| | - Andrey N Kuzmin
- Institute for Lasers, Photonics and Biophotonics and the Department of Chemistry, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Paras N Prasad
- Institute for Lasers, Photonics and Biophotonics and the Department of Chemistry, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - John-Bjarne Hansen
- Thrombosis Research Group, Institute of Clinical Medicine, Univesitet i Tromsø - The Arctic University of Norway, Tromsø, Norway; Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Olav Gaute Hellesø
- Department of Physics and Technology, Univesitet i Tromsø- The Arctic University of Norway, Tromsø, Norway
| | - Omri Snir
- Thrombosis Research Group, Institute of Clinical Medicine, Univesitet i Tromsø - The Arctic University of Norway, Tromsø, Norway; Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway.
| |
Collapse
|
16
|
Saffari PM, Asili P, Eshraghi S, Muhammadnejad A, Dehpour AR, Goudarzi R, Partoazar A. Phosphatidylserine accelerates wound healing and reduces necrosis in the rats: Growth factor activation. Clin Exp Pharmacol Physiol 2024; 51:e13849. [PMID: 38408759 DOI: 10.1111/1440-1681.13849] [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: 09/11/2023] [Revised: 12/20/2023] [Accepted: 01/30/2024] [Indexed: 02/28/2024]
Abstract
To examine the effect of topical phosphatidylserine (PS) on wound healing factors and tissue necrosis in in vivo models. Topical PS was applied to evaluate aspects of the wound healing process and growth factors production of vascular endothelial growth factors (VEGF) as well a necrosis reduction in the skin flap of rat models. Moreover, phenytoin (PHT) and cyclosporine A (CsA) were used topically as positive control treatments in wound and necrosis models, respectively. Immunohistochemistry (IHC) VEGF, transforming growth factor-β (TGF-β), fibroblast growth factor (FGF) and histopathology were analysed on the wounds of rats. In the necrosis assessment, necrotic areas were determined on photography taken from the back skin of rats. Results indicated that PS topically enhanced significantly (P < 0.05) numbers of fibroblasts and endothelium while inhibiting the neutrophils and macrophages during the 14 days of wound treatment. Moreover, higher values of collagen deposition and epithelialization scores as well as wound recovery percentage (near 80%) were determined significantly (P < 0.05) in the PS group compared with the control. IHC analysis determined that FGF and VEGF cytokine factors were elevated in the wound site by topical PS. Moreover, the necrotic area was significantly (P < 0.05) improved in the PS group. Our experiment indicated that wound improvement and flap survival values in PS treatments were superior to PHT and CsA control groups, respectively. In conclusion, these findings suggest the potential of PS application in the healing of wounds and control of necrosis development after surgery or skin injuries.
Collapse
Affiliation(s)
- Partow Mirzaee Saffari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Pooria Asili
- Department of Pathology, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadaf Eshraghi
- Department of Pharmaceutics, Faculty of Pharmacy, Islamic Azad University, Tehran, Iran
| | - Ahad Muhammadnejad
- Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Goudarzi
- Division of Research and Development, Pharmin USA, LLC, San Jose, California, USA
| | - Alireza Partoazar
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Weiss R, Mostageer M, Eichhorn T, Huber S, Egger D, Spittler A, Tripisciano C, Kasper C, Weber V. The fluorochrome-to-protein ratio is crucial for the flow cytometric detection of tissue factor on extracellular vesicles. Sci Rep 2024; 14:6419. [PMID: 38494537 PMCID: PMC10944842 DOI: 10.1038/s41598-024-56841-5] [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: 05/24/2023] [Accepted: 03/12/2024] [Indexed: 03/19/2024] Open
Abstract
Extracellular vesicles (EVs) have crucial roles in hemostasis and coagulation. They sustain coagulation by exposing phosphatidylserine and initiate clotting by surface expression of tissue factor (TF) under inflammatory conditions. As their relevance as biomarkers of coagulopathy is increasingly recognized, there is a need for the sensitive and reliable detection of TF+ EVs, but their flow cytometric analysis is challenging and has yielded controversial findings for TF expression on EVs in the vascular system. We investigated the effect of different fluorochrome-to-protein (F/P) ratios of anti-TF-fluorochrome conjugates on the flow cytometric detection of TF+ EVs from activated monocytes, mesenchymal stem cells (MSCs), and in COVID-19 plasma. Using a FITC-labeled anti-TF antibody (clone VD8), we show that the percentage of TF+ EVs declined with decreasing F/P ratios. TF was detected on 7.6%, 5.4%, and 1.1% of all EVs derived from activated monocytes at F/P ratios of 7.7:1, 6.6:1, and 5.2:1. A similar decline was observed for EVs from MSCs and for EVs in plasma, whereas the detection of TF on cells remained unaffected by different F/P ratios. We provide clear evidence that next to the antibody clone, the F/P ratio affects the flow cytometric detection of TF+ EVs and should be carefully controlled.
Collapse
Affiliation(s)
- René Weiss
- Center for Biomedical Technology, Department for Biomedical Research, University for Continuing Education Krems, Dr.-Karl-Dorrek-Strasse 30, 3500, Krems, Austria
| | - Marwa Mostageer
- Center for Biomedical Technology, Department for Biomedical Research, University for Continuing Education Krems, Dr.-Karl-Dorrek-Strasse 30, 3500, Krems, Austria
| | - Tanja Eichhorn
- Center for Biomedical Technology, Department for Biomedical Research, University for Continuing Education Krems, Dr.-Karl-Dorrek-Strasse 30, 3500, Krems, Austria
| | - Silke Huber
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Dominik Egger
- Institute of Cell and Tissue Culture Technology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Andreas Spittler
- Core Facility Flow Cytometry & Surgical Research Laboratories, Medical University of Vienna, Vienna, Austria
| | - Carla Tripisciano
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Cornelia Kasper
- Institute of Cell and Tissue Culture Technology, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Viktoria Weber
- Center for Biomedical Technology, Department for Biomedical Research, University for Continuing Education Krems, Dr.-Karl-Dorrek-Strasse 30, 3500, Krems, Austria.
| |
Collapse
|
18
|
Sacchetti S, Puricelli C, Mennuni M, Zanotti V, Giacomini L, Giordano M, Dianzani U, Patti G, Rolla R. Research into New Molecular Mechanisms in Thrombotic Diseases Paves the Way for Innovative Therapeutic Approaches. Int J Mol Sci 2024; 25:2523. [PMID: 38473772 DOI: 10.3390/ijms25052523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/12/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Thrombosis is a multifaceted process involving various molecular components, including the coagulation cascade, platelet activation, platelet-endothelial interaction, anticoagulant signaling pathways, inflammatory mediators, genetic factors and the involvement of various cells such as endothelial cells, platelets and leukocytes. A comprehensive understanding of the molecular signaling pathways and cell interactions that play a role in thrombosis is essential for the development of precise therapeutic strategies for the treatment and prevention of thrombotic diseases. Ongoing research in this field is constantly uncovering new molecular players and pathways that offer opportunities for more precise interventions in the clinical setting. These molecular insights into thrombosis form the basis for the development of targeted therapeutic approaches for the treatment and prevention of thrombotic disease. The aim of this review is to provide an overview of the pathogenesis of thrombosis and to explore new therapeutic options.
Collapse
Affiliation(s)
- Sara Sacchetti
- Clinical Chemistry Laboratory, "Maggiore della Carità" University Hospital, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
| | - Chiara Puricelli
- Clinical Chemistry Laboratory, "Maggiore della Carità" University Hospital, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
| | - Marco Mennuni
- Division of Cardiology, "Maggiore della Carità" University Hospital, Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy
| | - Valentina Zanotti
- Clinical Chemistry Laboratory, "Maggiore della Carità" University Hospital, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
| | - Luca Giacomini
- Clinical Chemistry Laboratory, "Maggiore della Carità" University Hospital, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
| | - Mara Giordano
- Clinical Chemistry Laboratory, "Maggiore della Carità" University Hospital, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
| | - Umberto Dianzani
- Clinical Chemistry Laboratory, "Maggiore della Carità" University Hospital, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
| | - Giuseppe Patti
- Division of Cardiology, "Maggiore della Carità" University Hospital, Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy
| | - Roberta Rolla
- Clinical Chemistry Laboratory, "Maggiore della Carità" University Hospital, Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
| |
Collapse
|
19
|
Pelzl L, Uzun G, Marini I, Zlamal J, Trumpp PN, Karakuyu A, Bakchoul T, Althaus K. Heparin-activated procoagulant platelet assay: a flow cytometry-based functional test for heparin-induced thrombocytopenia. J Thromb Haemost 2024; 22:470-479. [PMID: 37838242 DOI: 10.1016/j.jtha.2023.10.003] [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: 04/12/2023] [Revised: 09/04/2023] [Accepted: 10/05/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND Functional platelet activation assays are required for the diagnosis of heparin-induced thrombocytopenia (HIT). Due to their sophisticated methodology, they are only available in reference centers. OBJECTIVES To evaluate the diagnostic accuracy of the flow cytometry-based heparin-activated procoagulant platelet (HAPP) assay in the laboratory diagnosis of HIT. METHODS Procoagulant platelets (PCP), defined by the expression of phosphatidylserine and CD62-P, were evaluated by flow cytometry in platelet-rich plasma from healthy donors after incubation with patient sera in the absence and presence of heparin. A sample was considered positive in HAPP assay, if the following 3 criteria were met: 1) the percentage of PCPs was ≥10.3% after incubation with 0.2 IU/mL heparin, 2) the fold increase in presence of 0.2 IU/mL heparin compared with buffer was ≥1.5, and 3) 100 IU/mL of heparin resulted in ≥50% inhibition of PCP. HAPP assay was validated in a prospective cohort (n = 202) of consecutive specimens submitted to our laboratory for serologic diagnosis of HIT. Heparin-induced platelet activation (HIPA) assay was used as the reference standard. RESULTS HIT-positive sera induced PCPs in the presence of 0.2 IU/mL heparin, which was inhibited with 100 IU/mL of heparin. In the prospective validation cohort, there were 15 HIPA+ and 187 HIPA- sera. HAPP was positive in 20 samples in this cohort. Using optimized cut-offs, HAPP assay had a sensitivity of 93.3% and specificity of 96.8%. CONCLUSION HAPP assay is promising as a simple and reliable functional assay for HIT; however, further studies are needed to confirm our results in larger cohorts.
Collapse
Affiliation(s)
- Lisann Pelzl
- Institute for Clinical and Experimental Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany; Centre for Clinical Transfusion Medicine, Tübingen, Germany
| | - Günalp Uzun
- Institute for Clinical and Experimental Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany; Centre for Clinical Transfusion Medicine, Tübingen, Germany
| | - Irene Marini
- Institute for Clinical and Experimental Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany; Centre for Clinical Transfusion Medicine, Tübingen, Germany
| | - Jan Zlamal
- Institute for Clinical and Experimental Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany; Centre for Clinical Transfusion Medicine, Tübingen, Germany
| | - Pascal N Trumpp
- Institute for Clinical and Experimental Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Aleyna Karakuyu
- Institute for Clinical and Experimental Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Tamam Bakchoul
- Institute for Clinical and Experimental Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany; Centre for Clinical Transfusion Medicine, Tübingen, Germany.
| | - Karina Althaus
- Institute for Clinical and Experimental Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany; Centre for Clinical Transfusion Medicine, Tübingen, Germany
| |
Collapse
|
20
|
Ding B, Mao Y, Li Y, Xin M, Jiang S, Hu X, Xu Q, Ding Q, Wang X. A novel GATA1 variant p.G229D causing the defect of procoagulant platelet formation. Thromb Res 2024; 234:39-50. [PMID: 38159323 DOI: 10.1016/j.thromres.2023.12.015] [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: 06/05/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
INTRODUCTION GATA1 is one of the master transcription factors in hematopoietic lineages development which is crucial for megakaryocytic differentiation and maturation. Previous studies have shown that distinct GATA1 variants are associated with varying severities of macrothrombocytopenia and platelet dysfunction. OBJECTIVE To determine the underlying pathological mechanisms of a novel GATA1 variant (c. 686G > A, p. G229D) in a patient with recurrent traumatic muscle hematomas. METHODS Comprehensive phenotypic analysis of the patient platelets was performed. Procoagulant platelet formation and function were detected using flow cytometry assay and thrombin generation test (TGT), respectively. The ANO6 expression was measured by qPCR and western blot. The intracellular supramaximal calcium flux was detected by Fluo-5N fluorescent assay. RESULTS The patient displayed mild macrothrombocytopenia with defects of platelet granules, aggregation, and integrin αIIbβ3 activation. The percentage of the procoagulant platelet formation of the patient upon the stimulation of thrombin plus collagen was lower than that of the healthy controls (40.9 % vs 49.0 % ± 5.1 %). The patient platelets exhibited a marked reduction of thrombin generation in platelet rich plasma TGT compared to the healthy controls (peak value: ∼70 % of the healthy controls; the endogenous thrombin potential: ∼40 % of the healthy controls). The expression of ANO6 and intracellular calcium flux were impaired, which together with abnormal granules of the patient platelets might contribute to defect of procoagulant platelet function. CONCLUSIONS The G229D variant could lead to a novel platelet phenotype characterized by defective procoagulant platelet formation and function, which extended the range of GATA1 variants associated platelet disorders.
Collapse
Affiliation(s)
- Biying Ding
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yinqi Mao
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yang Li
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Min Xin
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shifeng Jiang
- State Key Laboratory of Microbial Metabolism & Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaobo Hu
- Department of Molecular Biology, Shanghai Center for Clinical Laboratory, Shanghai, China
| | - Qin Xu
- State Key Laboratory of Microbial Metabolism & Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Qiulan Ding
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Collaborative Innovation Center of Hematology, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Xuefeng Wang
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Collaborative Innovation Center of Hematology, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
21
|
Kumar S, Schroeder JA, Shi Q. Platelet-targeted gene therapy induces immune tolerance in hemophilia and beyond. J Thromb Haemost 2024; 22:23-34. [PMID: 37558132 PMCID: PMC11249137 DOI: 10.1016/j.jtha.2023.07.025] [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: 04/10/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 08/11/2023]
Abstract
Blood platelets have unique storage and delivery capabilities. Platelets play fundamental roles in hemostasis, inflammatory reactions, and immune responses. Beyond their functions, platelets have been used as a target for gene therapy. Platelet-targeted gene therapy aims to deliver a sustained expression of neo-protein in vivo by genetically modifying the target cells, resulting in a cure for the disease. Even though there has been substantial progress in the field of gene therapy, the potential development of immune responses to transgene products or vectors remains a significant concern. Of note, multiple preclinical studies using platelet-specific lentiviral gene delivery to hematopoietic stem cells in hemophilia have demonstrated promising results with therapeutic levels of neo-protein that rescue the hemorrhagic bleeding phenotype and induce antigen-specific immune tolerance. Further studies using ovalbumin as a surrogate protein for platelet gene therapy have shown robust antigen-specific immune tolerance induced via peripheral clonal deletions of antigen-specific CD4- and CD8-T effector cells and induction of antigen-specific regulatory T (Treg) cells. This review discusses platelet-targeted gene therapy, focusing on immune tolerance induction.
Collapse
Affiliation(s)
- Saurabh Kumar
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA
| | - Jocelyn A Schroeder
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA; Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Qizhen Shi
- Blood Research Institute, Versiti Wisconsin, Milwaukee, Wisconsin, USA; Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Children's Research Institute, Children's Wisconsin, Milwaukee, Wisconsin, USA; Midwest Athletes Against Childhood Cancer (MACC) Fund Research Center Milwaukee, Wisconsin, USA.
| |
Collapse
|
22
|
Al-Jipouri A, Eritja À, Bozic M. Unraveling the Multifaceted Roles of Extracellular Vesicles: Insights into Biology, Pharmacology, and Pharmaceutical Applications for Drug Delivery. Int J Mol Sci 2023; 25:485. [PMID: 38203656 PMCID: PMC10779093 DOI: 10.3390/ijms25010485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Extracellular vesicles (EVs) are nanoparticles released from various cell types that have emerged as powerful new therapeutic option for a variety of diseases. EVs are involved in the transmission of biological signals between cells and in the regulation of a variety of biological processes, highlighting them as potential novel targets/platforms for therapeutics intervention and/or delivery. Therefore, it is necessary to investigate new aspects of EVs' biogenesis, biodistribution, metabolism, and excretion as well as safety/compatibility of both unmodified and engineered EVs upon administration in different pharmaceutical dosage forms and delivery systems. In this review, we summarize the current knowledge of essential physiological and pathological roles of EVs in different organs and organ systems. We provide an overview regarding application of EVs as therapeutic targets, therapeutics, and drug delivery platforms. We also explore various approaches implemented over the years to improve the dosage of specific EV products for different administration routes.
Collapse
Affiliation(s)
- Ali Al-Jipouri
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
| | - Àuria Eritja
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| | - Milica Bozic
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| |
Collapse
|
23
|
Spurgeon BEJ, Frelinger AL. OMIP-097: High-parameter phenotyping of human platelets by spectral flow cytometry. Cytometry A 2023; 103:935-940. [PMID: 37786346 DOI: 10.1002/cyto.a.24797] [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: 04/18/2023] [Revised: 08/24/2023] [Accepted: 09/12/2023] [Indexed: 10/04/2023]
Abstract
Using spectral flow cytometry, we developed a 16-color panel for analysis of platelet phenotype and function in human whole blood. The panel contains markers of clinical relevance and follows an optimized protocol for the high-parameter phenotyping of (phosphatidylserine positive) procoagulant platelets. Inclusion of established markers, such as CD62P and PAC-1, allows the subsetting of classic (proinflammatory and proaggregatory) phenotypes, while addition of novel markers, such as TLR9, allows the resolution of platelets with nonclassic functions. Multiple inducible (C3b, CD63, CD107a, CD154, and TLT-1) and constitutive (CD29, CD31, CD32, CD36, CD42a, CD61, and GPVI) markers are also measurable, and we demonstrate the use of automatic gating for platelet analysis. The panel is widely applicable to research and clinical settings and can be readily modified, should users wish to tailor the panel to more specific needs.
Collapse
Affiliation(s)
- Benjamin E J Spurgeon
- Center for Platelet Research Studies, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew L Frelinger
- Center for Platelet Research Studies, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Das K, Keshava S, Mukherjee T, Wang J, Magisetty J, Kolesnick R, Pendurthi UR, Rao LVM. Factor VIIa releases phosphatidylserine-enriched extracellular vesicles from endothelial cells by activating acid sphingomyelinase. J Thromb Haemost 2023; 21:3414-3431. [PMID: 37875382 DOI: 10.1016/j.jtha.2023.08.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 08/05/2023] [Accepted: 08/23/2023] [Indexed: 10/26/2023]
Abstract
BACKGROUND Our recent studies showed that activated factor (F) VII (FVIIa) releases extracellular vesicles (EVs) from the endothelium. FVIIa-released EVs were found to be enriched with phosphatidylserine (PS) and contribute to the hemostatic effect of FVIIa in thrombocytopenia and hemophilia. OBJECTIVE To investigate mechanisms by which FVIIa induces EV biogenesis and enriches EVs with PS. METHODS FVIIa activation of acid sphingomyelinase (aSMase) was evaluated by its translocation to the cell surface. The role of aSMase in the biogenesis of FVIIa-induced EVs and their enrichment with PS was investigated using specific siRNAs and inhibitors of aSMase and its downstream metabolites. Wild-type and aSMase-/- mice were injected with a control vehicle or FVIIa. EVs released into circulation were quantified by nanoparticle tracking analysis. EVs hemostatic potential was assessed in a murine thrombocytopenia model. RESULTS FVIIa activation of aSMase is responsible for both the externalization of PS and the release of EVs in endothelial cells. FVIIa-induced aSMase activation led to ceramide generation and de novo expression of transmembrane protein 16F. Inhibitors of ceramidases, sphingosine kinase, or sphingosine-1-phosphate receptor modulator blocked FVIIa-induced expression of transmembrane protein 16F and PS externalization without interfering with FVIIa release of EVs. In vivo, FVIIa release of EVs was markedly impaired in aSMase-/- mice compared with wild-type mice. Administration of a low dose of FVIIa, sufficient to induce EVs release, corrected bleeding associated with thrombocytopenia in wild-type mice but not in aSMase-/- mice. CONCLUSION Our study identifies a novel mechanism by which FVIIa induces PS externalization and releases PS-enriched EVs.
Collapse
Affiliation(s)
- Kaushik Das
- Department of Cellular and Molecular Biology, UT Tyler School of Medicine, The University of Texas Health Science Center at Tyler, Tyler, TX, USA.
| | - Shiva Keshava
- Department of Cellular and Molecular Biology, UT Tyler School of Medicine, The University of Texas Health Science Center at Tyler, Tyler, TX, USA
| | - Tanmoy Mukherjee
- Department of Cellular and Molecular Biology, UT Tyler School of Medicine, The University of Texas Health Science Center at Tyler, Tyler, TX, USA
| | - Jue Wang
- Department of Cellular and Molecular Biology, UT Tyler School of Medicine, The University of Texas Health Science Center at Tyler, Tyler, TX, USA
| | - Jhansi Magisetty
- Department of Cellular and Molecular Biology, UT Tyler School of Medicine, The University of Texas Health Science Center at Tyler, Tyler, TX, USA
| | | | - Usha R Pendurthi
- Department of Cellular and Molecular Biology, UT Tyler School of Medicine, The University of Texas Health Science Center at Tyler, Tyler, TX, USA
| | - L Vijaya Mohan Rao
- Department of Cellular and Molecular Biology, UT Tyler School of Medicine, The University of Texas Health Science Center at Tyler, Tyler, TX, USA.
| |
Collapse
|
25
|
Gavioli G, Razzoli A, Bedolla DE, Di Bartolomeo E, Quartieri E, Iotti B, Berni P, Birarda G, Vaccari L, Schiroli D, Marraccini C, Baricchi R, Merolle L. Cryopreservation affects platelet macromolecular composition over time after thawing and differently impacts on cancer cells behavior in vitro. Platelets 2023; 34:2281943. [PMID: 38010129 DOI: 10.1080/09537104.2023.2281943] [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: 05/30/2023] [Accepted: 11/06/2023] [Indexed: 11/29/2023]
Abstract
Cryopreservation affects platelets' function, questioning their use for cancer patients. We aimed to investigate the biochemical events that occur over time after thawing to optimize transfusion timing and evaluate the effect of platelet supernatants on tumor cell behavior in vitro. We compared fresh (Fresh-PLT) with Cryopreserved platelets (Cryo-PLT) at 1 h, 3 h and 6 h after thawing. MCF-7 and HL-60 cells were cultured with Fresh- or 1 h Cryo-PLT supernatants to investigate cell proliferation, migration, and PLT-cell adhesion. We noticed a significant impairment of hemostatic activity accompanied by a post-thaw decrease of CD42b+ , which identifies the CD62P--population. FTIR spectroscopy revealed a decrease in the total protein content together with changes in their conformational structure, which identified two sub-groups: 1) Fresh and 1 h Cryo-PLT; 2) 3 h and 6 h cryo-PLT. Extracellular vesicle shedding and phosphatidylserine externalization (PS) increased after thawing. Cryo-PLT supernatants inhibited cell proliferation, impaired MCF-7 cell migration, and reduced ability to adhere to tumor cells. Within the first 3 hours after thawing, irreversible alterations of biomolecular structure occur in Cryo-PLT. Nevertheless, Cryo-PLT should be considered safe for the transfusion of cancer patients because of their insufficient capability to promote cancer cell proliferation, adhesion, or migration.
Collapse
Affiliation(s)
- Gaia Gavioli
- AUSL-IRCCS di Reggio Emilia, Transfusion Medicine Unit, Reggio Emilia, Italy
- Clinical and Experimental PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Agnese Razzoli
- AUSL-IRCCS di Reggio Emilia, Transfusion Medicine Unit, Reggio Emilia, Italy
- Clinical and Experimental PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Diana E Bedolla
- Elettra - Sincrotrone Trieste S.C.p.A, Basovizza, Italy
- Molecular Pathology Lab, International Center for Genetic Engineering and Biotechnology (ICGEB), Area Science Park, Trieste, Italy
- Center for Biospectroscopy and School of Chemistry, Monash University, Clayton, VIC, Australia
| | | | - Eleonora Quartieri
- AUSL-IRCCS di Reggio Emilia, Transfusion Medicine Unit, Reggio Emilia, Italy
| | - Barbara Iotti
- AUSL-IRCCS di Reggio Emilia, Transfusion Medicine Unit, Reggio Emilia, Italy
| | - Pamela Berni
- AUSL-IRCCS di Reggio Emilia, Transfusion Medicine Unit, Reggio Emilia, Italy
| | | | - Lisa Vaccari
- Elettra - Sincrotrone Trieste S.C.p.A, Basovizza, Italy
| | - Davide Schiroli
- AUSL-IRCCS di Reggio Emilia, Transfusion Medicine Unit, Reggio Emilia, Italy
| | - Chiara Marraccini
- AUSL-IRCCS di Reggio Emilia, Transfusion Medicine Unit, Reggio Emilia, Italy
| | - Roberto Baricchi
- AUSL-IRCCS di Reggio Emilia, Transfusion Medicine Unit, Reggio Emilia, Italy
| | - Lucia Merolle
- AUSL-IRCCS di Reggio Emilia, Transfusion Medicine Unit, Reggio Emilia, Italy
| |
Collapse
|
26
|
Zhang J, Hu X, Wang T, Xiao R, Zhu L, Ruiz M, Dupuis J, Hu Q. Extracellular vesicles in venous thromboembolism and pulmonary hypertension. J Nanobiotechnology 2023; 21:461. [PMID: 38037042 PMCID: PMC10691137 DOI: 10.1186/s12951-023-02216-3] [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: 08/17/2023] [Accepted: 11/15/2023] [Indexed: 12/02/2023] Open
Abstract
Venous thromboembolism (VTE) is a multifactorial disease, and pulmonary hypertension (PH) is a serious condition characterized by pulmonary vascular remodeling leading with increased pulmonary vascular resistance, ultimately leading to right heart failure and death. Although VTE and PH have distinct primary etiologies, they share some pathophysiologic similarities such as dysfunctional vasculature and thrombosis. In both conditions there is solid evidence that EVs derived from a variety of cell types including platelets, monocytes, endothelial cells and smooth muscle cells contribute to vascular endothelial dysfunction, inflammation, thrombosis, cellular activation and communications. However, the roles and importance of EVs substantially differ between studies depending on experimental conditions and parent cell origins of EVs that modify the nature of their cargo. Numerous studies have confirmed that EVs contribute to the pathophysiology of VTE and PH and increased levels of various EVs in relation with the severity of VTE and PH, confirming its potential pathophysiological role and its utility as a biomarker of disease severity and as potential therapeutic targets.
Collapse
Affiliation(s)
- Jiwei Zhang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China
- Department of Pathology, Union Hospital, Tongji Medical College, HUST, Wuhan, China
| | - Xiaoyi Hu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tao Wang
- Department of Respiratory Medicine, Tongji Hospital, Tongji Medical College, HUST, Wuhan, China
| | - Rui Xiao
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China
| | - Liping Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China
| | - Matthieu Ruiz
- Department of Nutrition, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montréal, Québec, Canada
| | - Jocelyn Dupuis
- Montreal Heart Institute, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Qinghua Hu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China.
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China.
| |
Collapse
|
27
|
Prouse T, Majumder R. TMEM16E: unscrambling our knowledge about coagulation. J Thromb Haemost 2023; 21:3000-3004. [PMID: 37633641 DOI: 10.1016/j.jtha.2023.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 08/28/2023]
Affiliation(s)
- Teagan Prouse
- Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Rinku Majumder
- Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| |
Collapse
|
28
|
Nair S, Nova-Lamperti E, Labarca G, Kulasinghe A, Short KR, Carrión F, Salomon C. Genomic communication via circulating extracellular vesicles and long-term health consequences of COVID-19. J Transl Med 2023; 21:709. [PMID: 37817137 PMCID: PMC10563316 DOI: 10.1186/s12967-023-04552-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/22/2023] [Indexed: 10/12/2023] Open
Abstract
COVID-19 continues to affect an unprecedented number of people with the emergence of new variants posing a serious challenge to global health. There is an expansion of knowledge in understanding the pathogenesis of Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and the impact of the acute disease on multiple organs. In addition, growing evidence reports that the impact of COVID-19 on different organs persists long after the recovery phase of the disease, leading to long-term consequences of COVID-19. These long-term consequences involve pulmonary as well as extra-pulmonary sequelae of the disease. Noteably, recent research has shown a potential association between COVID-19 and change in the molecular cargo of extracellular vesicles (EVs). EVs are vesicles released by cells and play an important role in cell communication by transfer of bioactive molecules between cells. Emerging evidence shows a strong link between EVs and their molecular cargo, and regulation of metabolism in health and disease. This review focuses on current knowledge about EVs and their potential role in COVID-19 pathogenesis, their current and future implications as tools for biomarker and therapeutic development and their possible effects on long-term impact of COVID-19.
Collapse
Affiliation(s)
- Soumyalekshmi Nair
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Qld, 4072, Australia
| | - Estefania Nova-Lamperti
- Molecular and Translational Immunology Laboratory, Clinical Biochemistry and Immunology Department, Pharmacy Faculty, Universidad de Concepción, Concepción, Chile
| | - Gonzalo Labarca
- Molecular and Translational Immunology Laboratory, Clinical Biochemistry and Immunology Department, Pharmacy Faculty, Universidad de Concepción, Concepción, Chile
| | - Arutha Kulasinghe
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Qld, 4102, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Flavio Carrión
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Qld, 4072, Australia.
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| |
Collapse
|
29
|
Hansen ES, Edvardsen MS, Aukrust P, Ueland T, Hansen JB, Brækkan SK, Morelli VM. Combined effect of high factor VIII levels and high mean platelet volume on the risk of future incident venous thromboembolism. J Thromb Haemost 2023; 21:2844-2853. [PMID: 37393000 DOI: 10.1016/j.jtha.2023.06.022] [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: 04/03/2023] [Revised: 06/10/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023]
Abstract
BACKGROUND High factor VIII (FVIII) levels and large platelets, as reflected by a high mean platelet volume (MPV), are separately associated with increased risk of venous thromboembolism (VTE). Whether the combination of high FVIII levels and large platelets has a supra-additive effect on VTE risk is unknown. OBJECTIVES We aimed to investigate the joint effect of high FVIII levels and large platelets, as reflected by high MPV, on the risk of future incident VTE. METHODS A population-based nested case-control study with 365 incident VTE cases and 710 controls was derived from the Tromsø study. FVIII antigen levels and MPV were measured in blood samples drawn at baseline. Odds ratios with 95% CIs were estimated across FVIII tertiles (<85%, 85%-108%, and ≥108%) and within predefined MPV strata (<8.5, 8.5-9.5, and ≥9.5 fL). RESULTS VTE risk increased linearly across FVIII tertiles (Ptrend < .001) in models adjusted for age, sex, body mass index, and C-reactive protein. In the combined analysis, participants with FVIII levels in the highest tertile and an MPV of ≥9.5 fL (ie, joint exposure) had an odds ratio for VTE of 2.71 (95% CI, 1.44-5.11) compared with those with FVIII levels in the lowest tertile and an MPV of <8.5 fL (reference). In the joint exposure group, 52% (95% CI, 17%-88%) of VTEs were attributable to the biological interaction between FVIII and MPV. CONCLUSION Our results suggest that large platelets, as reflected by high MPV, might play a role in the mechanism by which high FVIII level increases the risk of incident VTE.
Collapse
Affiliation(s)
- Ellen-Sofie Hansen
- Thrombosis Research Group, Department of Clinical Medicine, UiT-the Arctic University of Norway, Tromsø, Norway.
| | - Magnus S Edvardsen
- Thrombosis Research Group, Department of Clinical Medicine, UiT-the Arctic University of Norway, Tromsø, Norway
| | - Pål Aukrust
- Faculty of Medicine, University of Oslo, Oslo, Norway; Research Institute of Internal Medicine, Oslo, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Thor Ueland
- Thrombosis Research Group, Department of Clinical Medicine, UiT-the Arctic University of Norway, Tromsø, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway; Research Institute of Internal Medicine, Oslo, Norway
| | - John-Bjarne Hansen
- Thrombosis Research Group, Department of Clinical Medicine, UiT-the Arctic University of Norway, Tromsø, Norway; Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Sigrid K Brækkan
- Thrombosis Research Group, Department of Clinical Medicine, UiT-the Arctic University of Norway, Tromsø, Norway; Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Vânia M Morelli
- Thrombosis Research Group, Department of Clinical Medicine, UiT-the Arctic University of Norway, Tromsø, Norway; Thrombosis Research Center, Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
30
|
Zlamal J, Singh A, Weich K, Jaffal H, Uzun G, Pelzl L, Althaus K, Bakchoul T. Platelet phosphatidylserine is the critical mediator of thrombosis in heparin-induced thrombocytopenia. Haematologica 2023; 108:2690-2702. [PMID: 37102605 PMCID: PMC10542843 DOI: 10.3324/haematol.2022.282275] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 04/19/2023] [Indexed: 04/28/2023] Open
Abstract
Heparin-induced thrombocytopenia (HIT) is a severe immune-mediated prothrombotic disorder caused by antibodies (Ab) reactive to complexes of platelet factor 4 and heparin. Platelets (PLT) and their interaction with different immune cells contribute to prothrombotic conditions in HIT. However, the exact mechanisms and the role of different PLT subpopulations in this prothrombotic environment remain poorly understood. In this study, we observed that HIT patient Ab induce a new PLT population that is characterized by increased P-selectin expression and phosphatidylserine (PS) externalization. Formation of this procoagulant PLT subpopulation was dependent on engagement of PLT Fc-γ-RIIA by HIT Ab and resulted in a significant increase of thrombin generation on the PLT surface. Using an ex vivo thrombosis model and multi-parameter assessment of thrombus formation, we observed that HIT Ab-induced procoagulant PLT propagated formation of large PLT aggregates, leukocyte recruitment and most importantly, fibrin network generation. These prothrombotic conditions were prevented via the upregulation of PLT intracellular cAMP with Iloprost, a clinically approved prostacyclin analogue. Additionally, the functional relevance of P-selectin and PS was dissected. While inhibition of P-selectin did not affect thrombus formation, the specific blockade of PS prevented HIT Ab-mediated thrombin generation and most importantly procoagulant PLT-mediated thrombus formation ex vivo. Taken together, our findings indicate that procoagulant PLT are critical mediators of prothrombotic conditions in HIT. Specific PS targeting could be a promising therapeutic approach to prevent thromboembolic events in HIT patients.
Collapse
Affiliation(s)
- Jan Zlamal
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University of Tübingen, Tübingen, Germany; Centre for Clinical Transfusion Medicine, Tübingen
| | - Anurag Singh
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University of Tübingen, Tübingen
| | - Karoline Weich
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University of Tübingen, Tübingen
| | - Hisham Jaffal
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University of Tübingen, Tübingen
| | - Günalp Uzun
- Centre for Clinical Transfusion Medicine, Tübingen
| | - Lisann Pelzl
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University of Tübingen, Tübingen, Germany; Centre for Clinical Transfusion Medicine, Tübingen
| | - Karina Althaus
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University of Tübingen, Tübingen, Germany; Centre for Clinical Transfusion Medicine, Tübingen
| | - Tamam Bakchoul
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University of Tübingen, Tübingen, Germany; Centre for Clinical Transfusion Medicine, Tübingen.
| |
Collapse
|
31
|
Li M, Wu X, Shi J, Niu Y. Endothelium dysfunction and thrombosis in COVID-19 with type 2 diabetes. Endocrine 2023; 82:15-27. [PMID: 37392341 DOI: 10.1007/s12020-023-03439-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 06/21/2023] [Indexed: 07/03/2023]
Abstract
SARS-CoV-2 can directly or indirectly damage endothelial cells. Endothelial injury, especially phosphatidylserine (PS) exposure on the outer membrane of cells, can more easily promote thrombosis. Type 2 diabetes(T2D) patients were more susceptible to COVID-19, they had more severe symptoms, higher risk of thrombotic complications, and longer duration of post-COVID-19 sequelae. This review provided a detailed overview of the mechanisms underlying endothelial dysfunction in T2D patients with COVID-19 (including long COVID), which may be influenced by hyperglycemia, hypoxia, and pro-inflammatory environments. The mechanisms of thrombosis in T2D patients with COVID-19 are also explored, particularly the effects of increased numbers of PS-exposing particles, blood cells, and endothelial cells on hypercoagulability. Given the high risk of thrombosis in T2D patients with COVID-19, early antithrombotic therapy can both minimize the impact of the disease on patients and maximize the chances of improvement, thereby alleviating patient suffering. We provided detailed guidance on antithrombotic drugs and dosages for mild, moderate, and severe patients, emphasizing that the optimal timing of thromboprophylaxis is a critical factor in influencing prognosis. Considering the potential interactions between antidiabetic, anticoagulant, and antiviral drugs, we proposed practical and comprehensive management recommendations to supplement the incomplete efficacy of vaccines in the diabetic population, reduce the incidence of post-COVID-19 sequelae, and improve patient quality of life.
Collapse
Affiliation(s)
- Mengdi Li
- Department of Endodontics, The First Hospital, Harbin Medical University, Harbin, China
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, China
| | - Xiaoming Wu
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, China
| | - Jialan Shi
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, China
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yumei Niu
- Department of Endodontics, The First Hospital, Harbin Medical University, Harbin, China.
| |
Collapse
|
32
|
Lorusso A, Croxon H, Faherty-O'Donnell S, Field S, Fitzpatrick Á, Farrelly A, Hervig T, Waters A. The impact of donor biological variation on the quality and function of cold-stored platelets. Vox Sang 2023; 118:730-737. [PMID: 37439150 DOI: 10.1111/vox.13495] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/09/2023] [Accepted: 06/18/2023] [Indexed: 07/14/2023]
Abstract
BACKGROUND AND OBJECTIVES Room temperature-stored platelets (RTPs) maximize platelet viability but limit shelf life. The aims of this study were to investigate the impact of donor variability on cold-stored platelets (CSPs) and RTP, to determine whether RTP quality markers are appropriate for CSP. MATERIALS AND METHODS Double platelet donations (n = 10) were collected from consented regular male donors stored in 100% plasma. A full blood count, donor age, weight, height and body mass index (BMI) were collected at the time of donation. Platelet donations were split equally into two bags, and assigned to non-agitated CSP or agitated RTP. The quality and function of platelets were assessed throughout the standard 7 days of storage and at expiry (day 8). Non-parametric statistical analyses were used to analyse results given the small sample size. RESULTS As expected, there were significant differences between CSP and RTP throughout storage including a reduction in CSP concentration as well as a loss of swirling. Furthermore, a significant increase in CSP exhibiting activation and apoptotic markers was observed. Platelet concentrations were further impacted by donor BMI, and donors with the highest BMI (>29) had the lowest platelet concentration and activation response at the end of CSP storage. CONCLUSION Platelet quality and functionality play a vital role in transfusion outcomes; however, blood components are inherently variable. This study demonstrated, for the first time, the specific impact of donor BMI on CSP quality and function and highlights the requirement for novel quality markers for assessing CSPs.
Collapse
Affiliation(s)
- Alice Lorusso
- Irish Blood Transfusion Service, National Blood Centre, Dublin, Ireland
| | - Harry Croxon
- Irish Blood Transfusion Service, National Blood Centre, Dublin, Ireland
| | | | - Stephen Field
- Irish Blood Transfusion Service, National Blood Centre, Dublin, Ireland
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Áine Fitzpatrick
- Irish Blood Transfusion Service, National Blood Centre, Dublin, Ireland
| | - Aileen Farrelly
- Irish Blood Transfusion Service, National Blood Centre, Dublin, Ireland
| | - Tor Hervig
- Irish Blood Transfusion Service, National Blood Centre, Dublin, Ireland
| | - Allison Waters
- Irish Blood Transfusion Service, National Blood Centre, Dublin, Ireland
- UCD School of Public Health, Population Science and Physiotherapy, University College Dublin, Dublin, Ireland
| |
Collapse
|
33
|
Ballard-Kordeliski A, Lee RH, O'Shaughnessy EC, Kim PY, Jones S, Mackman N, Flick MJ, Paul DS, Adalsteinsson D, Bergmeier W. 4D intravital imaging studies identify platelets as the predominant cellular procoagulant surface in a mouse model of hemostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.25.554449. [PMID: 37662350 PMCID: PMC10473702 DOI: 10.1101/2023.08.25.554449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Interplay between platelets, coagulation/fibrinolytic factors, and endothelial cells (ECs) is necessary for effective hemostatic plug formation. This study describes a novel four-dimensional (4D) imaging platform to visualize and quantify hemostatic plug components with high spatiotemporal resolution. Fibrin accumulation following laser-induced endothelial ablation was observed at the EC-platelet plug interface, controlled by the antagonistic balance between fibrin generation and breakdown. Phosphatidylserine (PS) was first detected in close physical proximity to the fibrin ring, followed by exposure across the endothelium. Impaired PS exposure in cyclophilinD -/- mice resulted in a significant reduction in fibrin accumulation. Adoptive transfer and inhibitor studies demonstrated a key role for platelets, but not ECs, in fibrin generation during hemostatic plug formation. Inhibition of fibrinolysis with tranexamic acid (TXA) led to increased fibrin accumulation in WT mice, but not in cyclophilinD -/- mice or WT mice treated with antiplatelet drugs. These studies implicate platelets as the functionally dominant procoagulant surface during hemostatic plug formation. In addition, they suggest that impaired fibrin formation due to reduced platelet procoagulant activity is not reversed by TXA treatment.
Collapse
|
34
|
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: 5] [Impact Index Per Article: 2.5] [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
|
35
|
Das K, Paul S, Mukherjee T, Ghosh A, Sharma A, Shankar P, Gupta S, Keshava S, Parashar D. Beyond Macromolecules: Extracellular Vesicles as Regulators of Inflammatory Diseases. Cells 2023; 12:1963. [PMID: 37566042 PMCID: PMC10417494 DOI: 10.3390/cells12151963] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023] Open
Abstract
Inflammation is the defense mechanism of the immune system against harmful stimuli such as pathogens, toxic compounds, damaged cells, radiation, etc., and is characterized by tissue redness, swelling, heat generation, pain, and loss of tissue functions. Inflammation is essential in the recruitment of immune cells at the site of infection, which not only aids in the elimination of the cause, but also initiates the healing process. However, prolonged inflammation often brings about several chronic inflammatory disorders; hence, a balance between the pro- and anti-inflammatory responses is essential in order to eliminate the cause while producing the least damage to the host. A growing body of evidence indicates that extracellular vesicles (EVs) play a major role in cell-cell communication via the transfer of bioactive molecules in the form of proteins, lipids, DNA, RNAs, miRNAs, etc., between the cells. The present review provides a brief classification of the EVs followed by a detailed description of how EVs contribute to the pathogenesis of various inflammation-associated diseases and their implications as a therapeutic measure. The latter part of the review also highlights how EVs act as a bridging entity in blood coagulation disorders and associated inflammation. The findings illustrated in the present review may open a new therapeutic window to target EV-associated inflammatory responses, thereby minimizing the negative outcomes.
Collapse
Affiliation(s)
- Kaushik Das
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA
| | - Subhojit Paul
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India; (S.P.); (A.G.)
| | - Tanmoy Mukherjee
- School of Medicine, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA;
| | - Arnab Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India; (S.P.); (A.G.)
| | - Anshul Sharma
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA;
| | - Prem Shankar
- Department of Neurobiology, The University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA;
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura 281406, India;
| | - Shiva Keshava
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA
| | - Deepak Parashar
- Department of Medicine, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
36
|
Yong J, Abrams ST, Wang G, Toh CH. Cell-free histones and the cell-based model of coagulation. J Thromb Haemost 2023; 21:1724-1736. [PMID: 37116754 DOI: 10.1016/j.jtha.2023.04.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/06/2023] [Accepted: 04/20/2023] [Indexed: 04/30/2023]
Abstract
The cell-based model of coagulation remains the basis of our current understanding of clinical hemostasis and thrombosis. Its advancement on the coagulation cascade model has enabled new prohemostatic and anticoagulant treatments to be developed. In the past decade, there has been increasing evidence of the procoagulant properties of extracellular, cell-free histones (CFHs). Although high levels of circulating CFHs released following extensive cell death in acute critical illnesses, such as sepsis and trauma, have been associated with adverse coagulation outcomes, including disseminated intravascular coagulation, new information has also emerged on how its local effects contribute to physiological clot formation. CFHs initiate coagulation by tissue factor exposure, either by destruction of the endovascular barrier or induction of endoluminal tissue factor expression on endothelia and monocytes. CFHs can also bind prothrombin directly, generating thrombin via the alternative prothrombinase pathway. In amplifying and augmenting the procoagulant signal, CFHs activate and aggregate platelets, increase procoagulant material bioavailability through platelet degranulation and Weibel-Palade body exocytosis, activate intrinsic coagulation via platelet polyphosphate release, and induce phosphatidylserine exposure. CFHs also inhibit protein C activation and downregulate thrombomodulin expression to reduce anti-inflammatory and anticoagulant effects. In consolidating clot formation, CFHs augment the fibrin polymer to confer fibrinolytic resistance and integrate neutrophil extracellular traps into the clot structure. Such new information holds the promise of new therapeutic developments, including improved targeting of immunothrombotic pathologies in acute critical illnesses.
Collapse
Affiliation(s)
- Jun Yong
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK
| | - Simon T Abrams
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; Liverpool Clinical Laboratories, Liverpool, UK
| | - Guozheng Wang
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; Liverpool Clinical Laboratories, Liverpool, UK
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK; The Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
37
|
Bravo-Iñiguez CE, Fritz JR, Shukla S, Sarangi S, Thompson DA, Amin SG, Tsaava T, Chaudhry S, Valentino SP, Hoffman HB, Imossi CW, Addorisio ME, Valdes-Ferrer SI, Chavan SS, Blanc L, Czura CJ, Tracey KJ, Huston JM. Vagus nerve stimulation primes platelets and reduces bleeding in hemophilia A male mice. Nat Commun 2023; 14:3122. [PMID: 37264009 PMCID: PMC10235098 DOI: 10.1038/s41467-023-38505-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/05/2023] [Indexed: 06/03/2023] Open
Abstract
Deficiency of coagulation factor VIII in hemophilia A disrupts clotting and prolongs bleeding. While the current mainstay of therapy is infusion of factor VIII concentrates, inhibitor antibodies often render these ineffective. Because preclinical evidence shows electrical vagus nerve stimulation accelerates clotting to reduce hemorrhage without precipitating systemic thrombosis, we reasoned it might reduce bleeding in hemophilia A. Using two different male murine hemorrhage and thrombosis models, we show vagus nerve stimulation bypasses the factor VIII deficiency of hemophilia A to decrease bleeding and accelerate clotting. Vagus nerve stimulation targets acetylcholine-producing T lymphocytes in spleen and α7 nicotinic acetylcholine receptors (α7nAChR) on platelets to increase calcium uptake and enhance alpha granule release. Splenectomy or genetic deletion of T cells or α7nAChR abolishes vagal control of platelet activation, thrombus formation, and bleeding in male mice. Vagus nerve stimulation warrants clinical study as a therapy for coagulation disorders and surgical or traumatic bleeding.
Collapse
Affiliation(s)
- Carlos E Bravo-Iñiguez
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Jason R Fritz
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Shilpa Shukla
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Department of Pediatric Hematology and Oncology, Cohen Children's Medical Center, Northwell Health, Lake Success, NY, 11040, USA
| | - Susmita Sarangi
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Department of Pediatric Hematology and Oncology, Cohen Children's Medical Center, Northwell Health, Lake Success, NY, 11040, USA
| | - Dane A Thompson
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Department of Surgery, Northwell Health, 300 Community Drive, Manhasset, NY, 11030, USA
| | - Seema G Amin
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Department of Pediatric Hematology and Oncology, Cohen Children's Medical Center, Northwell Health, Lake Success, NY, 11040, USA
| | - Tea Tsaava
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Saher Chaudhry
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Sara P Valentino
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Hannah B Hoffman
- Department of Surgery, Northwell Health, 300 Community Drive, Manhasset, NY, 11030, USA
| | - Catherine W Imossi
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Meghan E Addorisio
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Sergio I Valdes-Ferrer
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Sangeeta S Chavan
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Lionel Blanc
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Departments of Molecular Medicine and Pediatrics, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Boulevard, Hempstead, NY, 11549, USA
| | - Christopher J Czura
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Kevin J Tracey
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Jared M Huston
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research at Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA.
- Department of Surgery, Northwell Health, 300 Community Drive, Manhasset, NY, 11030, USA.
- Department of Science Education, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Boulevard, Hempstead, NY, 11549, USA.
| |
Collapse
|
38
|
Gupta A, Marzook H, Ahmad F. Comorbidities and clinical complications associated with SARS-CoV-2 infection: an overview. Clin Exp Med 2023; 23:313-331. [PMID: 35362771 PMCID: PMC8972750 DOI: 10.1007/s10238-022-00821-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/12/2022] [Indexed: 01/08/2023]
Abstract
The novel severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) causes major challenges to the healthcare system. SARS-CoV-2 infection leads to millions of deaths worldwide and the mortality rate is found to be greatly associated with pre-existing clinical conditions. The existing dataset strongly suggests that cardiometabolic diseases including hypertension, coronary artery disease, diabetes and obesity serve as strong comorbidities in coronavirus disease (COVID-19). Studies have also shown the poor outcome of COVID-19 in patients associated with angiotensin-converting enzyme-2 polymorphism, cancer chemotherapy, chronic kidney disease, thyroid disorder, or coagulation dysfunction. A severe complication of COVID-19 is mostly seen in people with compromised medical history. SARS-CoV-2 appears to attack the respiratory system causing pneumonia, acute respiratory distress syndrome, which lead to induction of severe systemic inflammation, multi-organ dysfunction, and death mostly in the patients who are associated with pre-existing comorbidity factors. In this article, we highlighted the key comorbidities and a variety of clinical complications associated with COVID-19 for a better understanding of the etiopathogenesis of COVID-19.
Collapse
Affiliation(s)
- Anamika Gupta
- Cardiovascular Research Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, UAE
| | - Hezlin Marzook
- Cardiovascular Research Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, UAE
| | - Firdos Ahmad
- Cardiovascular Research Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, UAE.
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE.
| |
Collapse
|
39
|
Schmugge M, Franzoso FD, Winkler J, Kroiss S, Seiler M, Speer O, Rand ML. IVIg treatment increases thrombin activation of platelets and thrombin generation in paediatric patients with immune thrombocytopenia. Br J Haematol 2023; 201:1209-1219. [PMID: 36861460 DOI: 10.1111/bjh.18702] [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: 06/20/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 03/03/2023]
Abstract
Clinical manifestations and laboratory parameters of haemostasis were investigated in 23 children with newly diagnosed immune thrombocytopenia (ITP) before and after intravenous immunoglobulin (IVIg) treatment. ITP patients with platelet counts of less than 20 × 109 /L and mild bleeding symptoms, graded by a standardized bleeding score (BS), were compared with healthy children with normal platelet counts and children with chemotherapy-related thrombocytopenia. Markers of platelet activation and platelet apoptosis in the absence and presence of platelet activators were analysed by flow cytometry; thrombin generation in plasma was determined. ITP patients at diagnosis presented with increased proportions of platelets expressing CD62P and CD63 and activated caspases, and with decreased thrombin generation. Thrombin-induced activation of platelets was reduced in ITP compared with controls, while increased proportions of platelets with activated caspases were observed. Children with a higher BS had lower proportions of CD62P-expressing platelets compared with those with a lower BS. IVIg treatment increased the number of reticulated platelets, the platelet count to more than 20 × 109 /L and improved bleeding in all patients. Decreased thrombin-induced platelet activation, as well as thrombin generation, were ameliorated. Our results indicate that IVIg treatment helps to counteract diminished platelet function and coagulation in children with newly diagnosed ITP.
Collapse
Affiliation(s)
- Markus Schmugge
- Division of Hematology, Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Francesca Daniela Franzoso
- Division of Hematology, Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Jeannine Winkler
- Division of Hematology, Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Sabine Kroiss
- Division of Hematology Oncology, Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Monika Seiler
- Division of Hematology Emergency Department, Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Oliver Speer
- Center for Laboratory Medicine, Center for Laboratory Medicine, St. Gallen, Switzerland
| | - Margaret L Rand
- Division of Haematology/Oncology, Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
- Departments of Laboratory Medicine & Pathobiology, Biochemistry, and Paediatrics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
40
|
Platelet activation and ferroptosis mediated NETosis drives heme induced pulmonary thrombosis. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166688. [PMID: 36925054 DOI: 10.1016/j.bbadis.2023.166688] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023]
Abstract
Cell-free heme (CFH) is a product of hemoglobin, myoglobin and hemoprotein degradation, which is a hallmark of pathologies associated with extensive hemolysis and tissue damage. CHF and iron collectively induce cytokine storm, lung injury, respiratory distress and infection susceptibility in the lungs suggesting their key role in the progression of lung disease pathology. We have previously demonstrated that heme-mediated reactive oxygen species (ROS) induces platelet activation and ferroptosis. However, interaction of ferroptotic platelets and neutrophils, the mechanism of action and associated complications remain unclear. In this study, we demonstrate that heme-induced P-selectin expression and Phosphatidylserine (PS) externalization in platelets via ASK-1-inflammasome axis increases platelet-neutrophil aggregates in circulation, resulting in Neutrophil extracellular traps (NET) formation in vitro and in vivo. Further, heme-induced platelet activation in mice increased platelet-neutrophil aggregates and accumulation of NETs in the lungs causing pulmonary damage. Thus, connecting CFH-mediated platelet activation to NETosis and pulmonary thrombosis. As lung infections induce acute respiratory stress, thrombosis and NETosis, we propose that heme -mediated platelet activation and ferroptosis might be crucial in such clinical manifestations. Further, considering the ability of redox modulators and ferroptosis inhibitors like FS-1, Lpx-1 and DFO to inhibit heme-induced ferroptotic platelets-mediated NETosis and pulmonary thrombosis. They could be potential adjuvant therapy to regulate respiratory distress-associated clinical complications.
Collapse
|
41
|
Aslan JE. Fibrin reaches out to GPVI to influence how platelets shape clots. J Thromb Haemost 2023; 21:465-466. [PMID: 36858794 DOI: 10.1016/j.jtha.2022.11.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 03/02/2023]
Affiliation(s)
- Joseph E Aslan
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA; Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, USA; Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA.
| |
Collapse
|
42
|
Koltsova EM, Martyanov AA, Podoplelova NA. Procoagulant Properties of Extracellular Vesicles in Normal and Pathological Pregnancy. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES A: MEMBRANE AND CELL BIOLOGY 2023. [DOI: 10.1134/s1990747822060071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
43
|
Gui X, Chu X, Du Y, Wang Y, Zhang S, Ding Y, Tong H, Xu M, Li Y, Ju W, Sun Z, Li Z, Zeng L, Xu K, Qiao J. Impaired Platelet Function and Thrombus Formation in PDE5A-Deficient Mice. Thromb Haemost 2023; 123:207-218. [PMID: 36252813 DOI: 10.1055/a-1962-1613] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Intracellular cyclic GMP (cGMP) inhibits platelet function. Platelet cGMP levels are controlled by phosphodiesterase 5A (PDE5A)-mediated degradation. However, the exact role of PDE5A in platelet function and thrombus formation remains poorly understood. In this study, we characterized the role of PDE5A in platelet activation and function. Platelets were isolated from wild type or PDE5A-/- mice to measure platelet aggregation, activation, phosphatidylserine exposure (annexin-V binding), reactive oxygen species (ROS) generation, platelet spreading as well as clot retraction. Cytosolic calcium mobilization was measured using Fluo-4 AM by a microplate reader. Western blot was used to measure the phosphorylation of VASP, ERK1/2, p38, JNK, and AKT. FeCl3-induced arterial thrombosis and venous thrombosis were assessed to evaluate the in vivo hemostatic function and thrombus formation. Additionally, in vitro thrombus formation was assessed in a microfluidic whole-blood perfusion assay. PDE5A-deficient mice presented significantly prolonged tail bleeding time and delayed arterial and venous thrombus formation. PDE5A deficiency significantly inhibited platelet aggregation, ATP release, P-selectin expression, and integrin aIIbb3 activation. In addition, an impaired spreading on collagen or fibrinogen and clot retraction was observed in PDE5A-deficient platelets. Moreover, PDE5A deficiency reduced phosphatidylserine exposure, calcium mobilization, ROS production, and increased intracellular cGMP level along with elevated VASP phosphorylation and reduced phosphorylation of ERK1/2, p38, JNK, and AKT. In conclusion, PDE5A modulates platelet activation and function and thrombus formation, indicating that therapeutically targeting it might be beneficial for the treatment of thrombotic diseases.
Collapse
Affiliation(s)
- Xiang Gui
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Xiang Chu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Yuwei Du
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Yuhan Wang
- School of Medical Technology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Sixuan Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Yangyang Ding
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Huan Tong
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Mengdi Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Yue Li
- School of Medical Technology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Wen Ju
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Zengtian Sun
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Zhenyu Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China.,School of Medical Technology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, People's Republic of China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, People's Republic of China
| |
Collapse
|
44
|
Buerck JP, Foster KM, Larson PR, O'Rear EA. Shear stimulated red blood cell microparticles: Effect on clot structure, flow and fibrinolysis. Biorheology 2023; 59:43-59. [PMID: 36970891 DOI: 10.3233/bir-220012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
BACKGROUND Microparticles (MPs) have activity in thrombus promotion and generation. Erythrocyte microparticles (ErMPs) have been reported to accelerate fibrinolysis in the absence of permeation. We hypothesized that shear induced ErMPs would affect fibrin structure of clots and change flow with implications for fibrinolysis. OBJECTIVE To determine the effect of ErMPs on clot structure and fibrinolysis. METHODS Plasma with elevated ErMPs was isolated from whole blood or from washed red blood cells (RBCs) resuspended in platelet free plasma (PFP) after high shear. Dynamic light scattering (DLS) provided size distribution of ErMPs from sheared samples and unsheared PFP controls. Clots were formed by recalcification for flow/lysis experiments and examined by confocal microscopy and SEM. Flow rates through clots and time-to-lysis were recorded. A cellular automata model showed the effect of ErMPs on fibrin polymerization and clot structure. RESULTS Coverage of fibrin increased by 41% in clots formed from plasma of sheared RBCs in PFP over controls. Flow rate decreased by 46.7% under a pressure gradient of 10 mmHg/cm with reduction in time to lysis from 5.7 ± 0.7 min to 12.2 ± 1.1 min (p < 0.01). Particle size of ErMPs from sheared samples (200 nm) was comparable to endogenous microparticles. CONCLUSIONS ErMPs alter the fibrin network in a thrombus and affect hydraulic permeability resulting in decelerated delivery of fibrinolytic drugs.
Collapse
Affiliation(s)
- James P Buerck
- School of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, OK, USA
| | - Kylie M Foster
- School of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, OK, USA
| | - Preston R Larson
- Samuel Roberts Noble Electron Microscopy Laboratory, University of Oklahoma, Norman, OK, USA
| | - Edgar A O'Rear
- School of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, OK, USA
- Institute for Biomedical Engineering, Science and Technology, University of Oklahoma, Norman, OK, USA
| |
Collapse
|
45
|
Setua S, Thangaraju K, Dzieciatkowska M, Wilkerson RB, Nemkov T, Lamb DR, Tagaya Y, Boyer T, Rowden T, Doctor A, D'Alessandro A, Buehler PW. Coagulation potential and the integrated omics of extracellular vesicles from COVID-19 positive patient plasma. Sci Rep 2022; 12:22191. [PMID: 36564503 PMCID: PMC9780627 DOI: 10.1038/s41598-022-26473-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Extracellular vesicles (EVs) participate in cell-to-cell communication and contribute toward homeostasis under physiological conditions. But EVs can also contribute toward a wide array of pathophysiology like cancer, sepsis, sickle cell disease, and thrombotic disorders. COVID-19 infected patients are at an increased risk of aberrant coagulation, consistent with elevated circulating levels of ultra-high molecular weight VWF multimers, D-dimer and procoagulant EVs. The role of EVs in COVID-19 related hemostasis may depend on cells of origin, vesicular cargo and size, however this is not well defined. We hypothesized that the procoagulant potential of EV isolates from COVID-19 (+) patient plasmas could be defined by thrombin generation assays. Here we isolated small EVs (SEVs) and large EVs (LEVs) from hospitalized COVID-19 (+) patient (n = 21) and healthy donor (n = 20) plasmas. EVs were characterized by flow cytometry, Transmission electron microscopy, nanoparticle tracking analysis, plasma thrombin generation and a multi-omics approach to define coagulation potential. These data were consistent with differences in EV metabolite, lipid, and protein content when compared to healthy donor plasma isolated SEVs and LEVs. Taken together, the effect of EVs on plasma procoagulant potential as defined by thrombin generation and supported by multi-omics is enhanced in COVID-19. Further, we observe that this effect is driven both by EV size and phosphatidyl serine.
Collapse
Affiliation(s)
- Saini Setua
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kiruphagaran Thangaraju
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver-Anschutz Medical Campus, 12801 East 17th Ave., Aurora, CO, 80045, USA
| | - Rebecca B Wilkerson
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver-Anschutz Medical Campus, 12801 East 17th Ave., Aurora, CO, 80045, USA
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver-Anschutz Medical Campus, 12801 East 17th Ave., Aurora, CO, 80045, USA
| | - Derek R Lamb
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yutaka Tagaya
- Division of Virology, Pathogenesis and Cancer, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tori Boyer
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tobi Rowden
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Allan Doctor
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver-Anschutz Medical Campus, 12801 East 17th Ave., Aurora, CO, 80045, USA.
| | - Paul W Buehler
- Department of Pediatrics, Center for Blood Oxygen Transport and Hemostasis, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
46
|
Muravlev IA, Dobrovolsky AB, Antonova OA, Khaspekova SG, Mazurov AV. Effects of platelets activated by different agonists on fibrin formation and thrombin generation. Platelets 2022; 34:2139365. [DOI: 10.1080/09537104.2022.2139365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Ivan A. Muravlev
- National Medical Research Center of Cardiology, Russian Ministry of Health, Moscow, Russian Federation
| | - Anatoly B. Dobrovolsky
- National Medical Research Center of Cardiology, Russian Ministry of Health, Moscow, Russian Federation
| | - Olga A. Antonova
- National Medical Research Center of Cardiology, Russian Ministry of Health, Moscow, Russian Federation
| | - Svetlana G. Khaspekova
- National Medical Research Center of Cardiology, Russian Ministry of Health, Moscow, Russian Federation
| | - Alexey V. Mazurov
- National Medical Research Center of Cardiology, Russian Ministry of Health, Moscow, Russian Federation
| |
Collapse
|
47
|
Immunothrombosis and the Role of Platelets in Venous Thromboembolic Diseases. Int J Mol Sci 2022; 23:ijms232113176. [PMID: 36361963 PMCID: PMC9656618 DOI: 10.3390/ijms232113176] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 12/05/2022] Open
Abstract
Venous thromboembolism (VTE) is the third leading cardiovascular cause of death and is conventionally treated with anticoagulants that directly antagonize coagulation. However, recent data have demonstrated that also platelets play a crucial role in VTE pathophysiology. In the current review, we outline how platelets are involved during all stages of experimental venous thrombosis. Platelets mediate initiation of the disease by attaching to the vessel wall upon which they mediate leukocyte recruitment. This process is referred to as immunothrombosis, and within this novel concept inflammatory cells such as leukocytes and platelets directly drive the progression of VTE. In addition to their involvement in immunothrombosis, activated platelets can directly drive venous thrombosis by supporting coagulation and secreting procoagulant factors. Furthermore, fibrinolysis and vessel resolution are (partly) mediated by platelets. Finally, we summarize how conventional antiplatelet therapy can prevent experimental venous thrombosis and impacts (recurrent) VTE in humans.
Collapse
|
48
|
Uzun G, Singh A, Abou-Khalel W, Pelzl L, Weich K, Nowak-Harnau S, Althaus K, Bugert P, Klüter H, Bakchoul T. Platelets and Sera from Donors of Convalescent Plasma after Mild COVID-19 Show No Procoagulant Phenotype. Hamostaseologie 2022; 42:S14-S23. [DOI: 10.1055/a-1797-0564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023] Open
Abstract
AbstractCoronavirus disease-2019 (COVID-19) is associated with increased thromboembolic complications. Long-term alteration in the coagulation system after acute COVID-19 infection is still a subject of research. Furthermore, the effect of sera from convalescent subjects on platelets is not known. In this study, we investigated platelet phenotype, coagulation, and fibrinolysis in COVID-19 convalescent plasma (CCP) donors and analyzed convalescent sera-induced effects on platelets. We investigated CCP donors who had a history of mild COVID-19 infection and donors who did not have COVID-19 were used as controls. We analyzed phosphatidylserine (PS) externalization, CD62p expression, and glycoprotein VI (GPVI) shedding both in platelet-rich plasma (PRP) and after incubation of washed healthy platelets with donors' sera using flow cytometry. Coagulation and fibrinolysis systems were assessed with thromboelastometry. Forty-seven CCP donors (22 males, 25 females; mean age (±SD): 41.4 ± 13.7 years) with a history of mild COVID-19 infection were included. Median duration after acute COVID-19 infection was 97 days (range, 34–401). We did not find an increased PS externalization, CD62p expression, or GPVI shedding in platelets from CCP donors. Sera from CCP donors did not induce PS externalization or GPVI shedding in healthy platelets. Sera-induced CD62p expression was slightly, albeit statistically significantly, lower in CCP donors than in plasma donors without a history of COVID-19. One patient showed increased maximum clot firmness and prolonged lysis time in thromboelastometry. Our findings suggest that procoagulant platelet phenotype is not present after mild COVID-19. Furthermore, CCP sera do not affect the activation status of platelets.
Collapse
Affiliation(s)
- Günalp Uzun
- Centre for Clinical Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Anurag Singh
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University Hospital of Tübingen, Tübingen, Germany
| | - Wissam Abou-Khalel
- Centre for Clinical Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Lisann Pelzl
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University Hospital of Tübingen, Tübingen, Germany
| | - Karoline Weich
- Centre for Clinical Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Stefanie Nowak-Harnau
- Centre for Clinical Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Karina Althaus
- Centre for Clinical Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University Hospital of Tübingen, Tübingen, Germany
| | - Peter Bugert
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Hessen, Mannheim, Germany
| | - Harald Klüter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Hessen, Mannheim, Germany
| | - Tamam Bakchoul
- Centre for Clinical Transfusion Medicine, University Hospital of Tübingen, Tübingen, Germany
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, University Hospital of Tübingen, Tübingen, Germany
| |
Collapse
|
49
|
Bourguignon A, Tasneem S, Hayward CPM. Update on platelet procoagulant mechanisms in health and in bleeding disorders. Int J Lab Hematol 2022; 44 Suppl 1:89-100. [PMID: 36074709 DOI: 10.1111/ijlh.13866] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/20/2022] [Indexed: 11/28/2022]
Abstract
Platelet procoagulant mechanisms are emerging to be complex and important to achieving haemostasis. The mechanisms include the release of procoagulant molecules from platelet storage granules, and strong agonist-induced expression of procoagulant phospholipids on the outer platelet membrane for tenase and prothrombinase assembly. The release of dense granule polyphosphate is important to platelet procoagulant function as it promotes the activation of factors XII, XI and V, inhibits tissue factor pathway inhibitor and fibrinolysis, and strengthens fibrin clots. Platelet procoagulant function also involves the release of partially activated factor V from platelets. Scott syndrome has provided important insights on the mechanisms that regulate procoagulant phospholipids expression on the external platelet membrane, which require strong agonist stimulation that increase cystolic calcium levels, mitochondrial calcium uptake, the loss of flippase function and activation of the transmembrane scramblase protein anoctamin 6. There have been advances in the methods used to directly and indirectly assess platelet procoagulant function in health and disease. Assessments of thrombin generation with platelet rich plasma samples has provided new insights on how platelet procoagulant function is altered in inherited platelet disorders, and how platelets influence the bleeding phenotype of a number of severe coagulation factor deficiencies. Several therapies, including desmopressin and recombinant factor VIIa, improve thrombin generation by platelets. There is growing interest in targeting platelet procoagulant function for therapeutic benefit. This review highlights recent advances in our understanding of platelet-dependent procoagulant mechanisms in health and in bleeding disorders.
Collapse
Affiliation(s)
- Alex Bourguignon
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada.,Hamilton Regional Laboratory Medicine Program, Hamilton, Canada
| | - Subia Tasneem
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Catherine P M Hayward
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada.,Hamilton Regional Laboratory Medicine Program, Hamilton, Canada.,Department of Medicine, McMaster University, Hamilton, Canada
| |
Collapse
|
50
|
Ye SL, Li WD, Li WX, Xiao L, Ran F, Chen MM, Li XQ, Sun LL. The regulatory role of exosomes in venous thromboembolism. Front Cell Dev Biol 2022; 10:956880. [PMID: 36092737 PMCID: PMC9449368 DOI: 10.3389/fcell.2022.956880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022] Open
Abstract
Exosomes are nanoscale endocytic vesicles, 30-150 nm in diameter, secreted by most cells. They mainly originate from multivesicular bodies formed by intracellular invagination of lysosomal microparticles, and released into the extracellular matrix after fusion of multivesicular bodies with cell membrane. Studies have shown that exosomes contain a variety of active molecules, such as proteins, lipids and RNAs (such as mRNA, miRNA, lncRNA, circRNA, etc.), which regulate the behavior of recipient cells and serve as circulating biomarkers of diseases, including thrombosis. Therefore, exosome research is important for the diagnosis, treatment, therapeutic monitoring, and prognosis of thrombosis in that it can reveal the counts, surface marker expression, protein, and miRNA cargo involved. Recent studies have shown that exosomes can be used as therapeutic vectors for tissue regeneration and as alternative vectors for drug delivery. In this review, we summarize the physiological and biochemical characteristics, isolation, and identification of exosomes. Moreover, we focus on the role of exosomes in thrombosis, specifically venous thromboembolism, and their potential clinical applications, including as biomarkers and therapeutic vectors for thrombosis.
Collapse
Affiliation(s)
- Sheng-Lin Ye
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wen-Dong Li
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wei-Xiao Li
- Department of Vascular Surgery, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Lun Xiao
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Feng Ran
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Meng-Meng Chen
- School of Electronic Engineering, Nanjing Xiaozhuang University, Nanjing, China
| | - Xiao-Qiang Li
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Li-Li Sun
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|