1
|
Bechtel GN, Sugerman GP, Eades T, Malinowska Z, Bush AM, Saber H, Parekh SH, Rausch MK. Mechano-Lysis in Whole Blood Clots: On How Mechanics Affect Clot Lysis, and How Lysis Affects Clot Mechanics. Adv Healthc Mater 2025; 14:e2403389. [PMID: 39981775 DOI: 10.1002/adhm.202403389] [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: 09/06/2024] [Revised: 02/05/2025] [Indexed: 02/22/2025]
Abstract
Thromboembolic diseases are a significant cause of mortality and are clinically treated enzymatically with tissue plasminogen activator (tPA). Interestingly, prior studies in fibrin fibers and fibrin gels have demonstrated that thrombolysis may be mechanically sensitive. This study aims to expand mechano-lytic studies to whole blood clots. Furthermore, this study investigates not only how mechanics impacts lysis but also how lysis impacts mechanics. Therefore, clots made from whole human blood are exposed to tPA while the clots are either stretched or unstretched. After, the resulting degree of clot lysis is measured by weighing the clots and by measuring the concentration of D-dimer in the surrounding bath. Additionally, each clot's mechanical properties are measured. This study finds that mechanical stretch accelerates loss in clot weight but does not impact the lysis rate as measured by D-dimer. Moreover, lysis not only removes clot volume but also reduces the remaining clot's stiffness and toughness. In summary, tPA-induced lysis of whole clot appears mechanically insensitive, but stretch reduces clot weight. Furthermore, results show that thrombolysis weakens clot. This suggests that thrombolysis may increase the risk of secondary embolizations but may also ease clot removal during thrombectomy, for example.
Collapse
Affiliation(s)
- Grace N Bechtel
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Gabriella P Sugerman
- Department of Biomedical Engineering, San José State University, San Jose, CA, 95192, USA
| | - Tatum Eades
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Zuzanna Malinowska
- Department of Aerospace Engineering & Engineering Mechanics, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Adam M Bush
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Hamidreza Saber
- Department of Neurology and Neurosurgery, Dell Medical School, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Sapun H Parekh
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Manuel K Rausch
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
- Department of Aerospace Engineering & Engineering Mechanics, The University of Texas at Austin, Austin, TX, 78712, USA
- Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
2
|
Khismatullin RR, Khabirova AI, Saliakhutdinova SM, Litvinov RI, Weisel JW. Shapes and Dimensions of Blood Clots Affect the Rate and Extent of Platelet-Driven Clot Contraction and Will Determine the Outcomes of Thrombosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.30.646236. [PMID: 40236199 PMCID: PMC11996312 DOI: 10.1101/2025.03.30.646236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Background Contraction (retraction, shrinkage) of hemostatic blood clots and obstructive thrombi is an important pathophysiological process. To mimic thrombi of various locations, we sought to determine whether the initial shape and dimensions of blood clots affect the rate and extent of clot contraction. Methods Thrombin-induced clots were formed in 0.3-5.7 ml samples of citrated human blood or platelet-rich plasma in a cylinder, cuboid, or flat chamber. The clots were allowed to contract at 37°C for 60 minutes with shrinkage tracked photographically. Following complete contraction, the physiologically most relevant cylindrical clots of various initial volumes (0.5 ml or 1.5 ml) were analyzed with scanning electron microscopy for composition and spatial non-uniformity with the emphasis on compressed polyhedral erythrocytes (polyhedrocytes). Results With the same volumes studied, the rates and final extents of contraction of whole blood clots were shape-dependent in the following order: flat > cuboid = cylindrical. Irrespective of the shape, the initially smaller clots always contracted to a larger extent. Unlike clots in whole blood, the platelet-rich plasma clots contracted almost independently of the clot volumes and shapes studied, indicating a key role of erythrocytes. The smaller blood clots with a higher extent of contraction contained more relative volume fraction of erythrocytes, especially compressed polyhedrocytes, due to tight packing and a decrease in the intercellular space. Unlike the smaller clots, the larger clots were not clearly segregated into distinct layers, reflecting incomplete spatial redistribution of blood clot components typical for contraction. Conclusions Contraction of blood clots depends on their shape and size. The smaller and larger clots have distinct size-dependent rates and extents of contraction as well as degrees of structural non-uniformity, reflecting different spatial gradients of compressive stresses. The physiological relevance of these findings is related to the variable geometry and size of intravascular blood clots and thrombi.
Collapse
|
3
|
Weisel JW, Litvinov RI. Ubiquitous clot contraction blockers hit ubiquitin and more. Blood Adv 2025; 9:1092-1094. [PMID: 40019777 PMCID: PMC11938070 DOI: 10.1182/bloodadvances.2024014973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025] Open
Affiliation(s)
- John W Weisel
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Rustem I Litvinov
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
4
|
Zaldua JC, Watson O, Gregoire DJ, Pillai S, Hellsten Y, Hawkins K, Evans PA. The impact of physical activity and intensity on clot mechanical microstructure and contraction in middle-aged/older habitual runners. BMC Neurol 2025; 25:81. [PMID: 40025415 PMCID: PMC11871672 DOI: 10.1186/s12883-025-04074-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] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 02/06/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Exercise in healthy individuals is associated with a hypercoagulable phase, leading to a temporary increase in clot mass and strength, which are controlled by an effective fibrinolytic system. Conversely, people with cardiovascular diseases often have a reduced fibrinolytic pathway, increased clot mass and abnormal clot contraction, resulting in poorer outcomes. We assessed clot microstructure, particularly the contractile forces of clot formation, in response to two exercise intensities in middle-aged/older runners. METHODS Twenty-eight habitual male and female runners aged over 40 years completed a 10 km moderate-intensity run; 14 of them performed a 3 km high-intensity run. Blood samples were collected at baseline, immediately postexercise and after 1 h of rest. Clot structural biomarkers df, gel time, and measurements of mature clot mechanical properties (gel time, G'Max and CFmax) were analysed alongside conventional plasma markers. RESULTS Both exercise intensities altered markers of coagulant activity (PT, APTT and FVIII) and fibrinolysis (D-dimer), indicating hypercoagulability. Compared with longer-duration lower-intensity exercise, df was greater after short-duration intensified exercise bouts. Following an hour of rest, df dropped to baseline levels. Additionally, CFmax decreased across timepoints at both exercise intensities. This effect was noted after one hour of rest compared with baseline, suggesting continuous fibrinolytic activity postexercise. CONCLUSION Exercise transiently induces an intensity-dependent hypercoagulable state, resulting in denser clot formation and a reduced clot contractile force due to fibrinolysis. These findings can help guide the safe commencement of rehabilitation exercise programs for cerebrovascular patients.
Collapse
Affiliation(s)
- J C Zaldua
- The Welsh Centre for Emergency Medicine Research, Emergency Department Morriston Hospital, Swansea Bay University Health Board, Swansea, Wales, SA6 6NL, UK
- Faculty of Medicine and Life Health Sciences, Medical School, Swansea University, Swansea, SA6 6NL, UK
| | - O Watson
- The Welsh Centre for Emergency Medicine Research, Emergency Department Morriston Hospital, Swansea Bay University Health Board, Swansea, Wales, SA6 6NL, UK
| | - D J Gregoire
- The Welsh Centre for Emergency Medicine Research, Emergency Department Morriston Hospital, Swansea Bay University Health Board, Swansea, Wales, SA6 6NL, UK
| | - S Pillai
- The Welsh Centre for Emergency Medicine Research, Emergency Department Morriston Hospital, Swansea Bay University Health Board, Swansea, Wales, SA6 6NL, UK
- Faculty of Medicine and Life Health Sciences, Medical School, Swansea University, Swansea, SA6 6NL, UK
| | - Y Hellsten
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - K Hawkins
- Faculty of Medicine and Life Health Sciences, Medical School, Swansea University, Swansea, SA6 6NL, UK
| | - P A Evans
- The Welsh Centre for Emergency Medicine Research, Emergency Department Morriston Hospital, Swansea Bay University Health Board, Swansea, Wales, SA6 6NL, UK.
- Faculty of Medicine and Life Health Sciences, Medical School, Swansea University, Swansea, SA6 6NL, UK.
| |
Collapse
|
5
|
Nurden AT, Nurden P. Glanzmann Thrombasthenia 10 Years Later: Progress Made and Future Directions. Semin Thromb Hemost 2025; 51:196-208. [PMID: 38499192 DOI: 10.1055/s-0044-1782519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Glanzmann thrombasthenia (GT) is the most common inherited platelet disorder (IPD) with mucocutaneous bleeding and a failure of platelets to aggregate when stimulated. The molecular cause is insufficient or defective αIIbβ3, an integrin encoded by the ITGA2B and ITGB3 genes. On activation αIIbβ3 undergoes conformational changes and binds fibrinogen (Fg) and other proteins to join platelets in the aggregate. The application of next-generation sequencing (NGS) to patients with IPDs has accelerated genotyping for GT; progress accompanied by improved mutation curation. The evaluation by NGS of variants in other hemostasis and vascular genes is a major step toward understanding why bleeding varies so much between patients. The recently discovered role for glycoprotein VI in thrombus formation, through its binding to fibrin and surface-bound Fg, may offer a mechanosensitive back-up for αIIbβ3, especially at sites of inflammation. The setting up of national networks for IPDs and GT is improving patient care. Hematopoietic stem cell therapy provides a long-term cure for severe cases; however, prophylaxis by monoclonal antibodies designed to accelerate fibrin formation at injured sites in the vasculature is a promising development. Gene therapy using lentil-virus vectors remains a future option with CRISPR/Cas9 technologies offering a promising alternative route.
Collapse
Affiliation(s)
- Alan T Nurden
- Institut Hospitalo-Universitaire LIRYC, Hôpital Xavier Arnozan, Pessac, France
| | - Paquita Nurden
- Institut Hospitalo-Universitaire LIRYC, Hôpital Xavier Arnozan, Pessac, France
| |
Collapse
|
6
|
Kliuchnikov E, Peshkova AD, Vo MQ, Marx KA, Litvinov RI, Weisel JW, Purohit PK, Barsegov V. Exploring effects of platelet contractility on the kinetics, thermodynamics, and mechanisms of fibrin clot contraction. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:6. [PMID: 40012560 PMCID: PMC11850289 DOI: 10.1038/s44341-025-00011-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/22/2025] [Indexed: 02/28/2025]
Abstract
Mechanisms of blood clot contraction - platelet-driven fibrin network remodeling, are not fully understood. We developed a detailed computational ClotDynaMo model of fibrin network with activated platelets, whose clot contraction rate for normal 450,000/µl human platelets depends on serum viscosity η, platelet filopodia length l, and weakly depends on filopodia traction force f and filopodia extension-retraction speed v. Final clot volume is independent of η, but depends on v, f and l. Analysis of ClotDynaMo output revealed a 2.24 TJ/mol clot contraction free energy change, with ~67% entropy and ~33% internal energy changes. The results illuminate the "optimal contraction principle" that maximizes volume change while minimizing energy cost. An 8-chain continuum model of polymer elasticity containing platelet forces, captures clot contractility as a function of platelet count, η and l. The ClotDynaMo and continuum models can be extended to include red blood cells, variable platelet properties, and mechanics of fibrin network.
Collapse
Affiliation(s)
| | - Alina D. Peshkova
- Departments of Pharmacology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA USA
| | - Minh Quan Vo
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia, PA USA
| | - Kenneth A. Marx
- Department of Chemistry, University of Massachusetts, Lowell, MA USA
| | - Rustem I. Litvinov
- Departments of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA USA
| | - John W. Weisel
- Departments of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA USA
| | - Prashant K. Purohit
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia, PA USA
| | - Valeri Barsegov
- Department of Chemistry, University of Massachusetts, Lowell, MA USA
| |
Collapse
|
7
|
Risman RA, Sen M, Tutwiler V, Hudson NE. Deconstructing fibrin(ogen) structure. J Thromb Haemost 2025; 23:368-380. [PMID: 39536819 PMCID: PMC11786978 DOI: 10.1016/j.jtha.2024.10.024] [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: 05/03/2024] [Revised: 09/18/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Fibrinogen and its insoluble degradation product fibrin are pivotal plasma proteins that play important roles in blood coagulation, wound healing, and immune responses. This review highlights research from the last 24 months connecting our progressing view of fibrin(ogen)'s structure, and in particular its conformational flexibility and posttranslational modifications, to its (patho)physiologic roles, molecular interactions, mechanical properties, use as a biomaterial, and potential as a therapeutic target. Recent work suggests that fibrinogen structure is highly dynamic, sampling multiple conformations, which may explain its myriad physiologic functions and the presence of cryptic binding sites. Investigations into fibrin clot structure elucidated the impact of posttranslational modifications, therapeutic interventions, and pathologic conditions on fibrin network morphology, offering insights into thrombus formation and embolization. Studies exploring the mechanical properties of fibrin reveal its response to blood flow and platelet-driven contraction, offering implications for clot stability and embolization risk. Moreover, advancements in tissue engineering leverage fibrin's biocompatibility and customizable properties for diverse applications, from wound healing to tissue regeneration and biomaterial interactions. These findings underscore the structural origins of fibrin(ogen)'s multifaceted roles and its potential as a target for therapeutic interventions.
Collapse
Affiliation(s)
- Rebecca A Risman
- Department of Biomedical Engineering, Rutgers University, New Brunswick, New Jersey, USA. https://twitter.com/rebecca_risman
| | - Mehmet Sen
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Valerie Tutwiler
- Department of Biomedical Engineering, Rutgers University, New Brunswick, New Jersey, USA. https://twitter.com/vatutwiler
| | - Nathan E Hudson
- Department of Physics, East Carolina University, Greenville, North Carolina, USA.
| |
Collapse
|
8
|
He W, Ding F, Zhang L, Liu W. In situ osteogenic activation of mesenchymal stem cells by the blood clot biomimetic mechanical microenvironment. Nat Commun 2025; 16:1162. [PMID: 39880808 PMCID: PMC11779924 DOI: 10.1038/s41467-025-56513-6] [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: 04/30/2024] [Accepted: 01/21/2025] [Indexed: 01/31/2025] Open
Abstract
Blood clots (BCs) play a crucial biomechanical role in promoting osteogenesis and regulating mesenchymal stem cell (MSC) function and fate. This study shows that BC formation enhances MSC osteogenesis by activating Itgb1/Fak-mediated focal adhesion and subsequent Runx2-mediated bone regeneration. Notably, BC viscoelasticity regulates this effect by modulating Runx2 nuclear translocation. To mimic this property, a viscoelastic peptide bionic hydrogel named BCgel was developed, featuring a nanofiber network, Itgb1 binding affinity, BC-like viscoelasticity, and biosafety. The anticipated efficacy of BCgel is demonstrated by its ability to induce nuclear translocation of Runx2 and promote bone regeneration in both in vitro experiments and in vivo bone defect models with blood clot defect, conducted on rats as well as beagles. This study offers insights into the mechano-transduction mechanisms of MSCs during osteogenesis and presents potential guidelines for the design of viscoelastic hydrogels in bone regenerative medicine.
Collapse
Affiliation(s)
- Wangxiao He
- Department of Stomatology, Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Xi'an, China.
- Department of Medical Oncology and Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, P. R. China.
| | - Fan Ding
- Department of Stomatology, Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Xi'an, China
| | - Liqiang Zhang
- Department of Stomatology, Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Xi'an, China
| | - Wenjia Liu
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Xi'an, China.
- Department of Medical Oncology and Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, P. R. China.
| |
Collapse
|
9
|
Del Carpio-Cano F, Songdej N, Guan L, Mao G, Goldfinger LE, Wurtzel JG, Lee K, Lambert MP, Poncz M, Rao AK. Transcription factor RUNX1 regulates coagulation factor XIII-A ( F13A1): decreased platelet-megakaryocyte F13A1 expression and clot contraction in RUNX1 haplodeficiency. Res Pract Thromb Haemost 2025; 9:102680. [PMID: 39995753 PMCID: PMC11849627 DOI: 10.1016/j.rpth.2025.102680] [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: 10/23/2024] [Revised: 12/13/2024] [Accepted: 01/09/2025] [Indexed: 02/26/2025] Open
Abstract
Background Germline RUNX1 haplodeficiency (RHD) is associated with thrombocytopenia, platelet dysfunction, and predisposition to myeloid malignancies. Platelet expression profiling of an RHD patient showed decreased F13A1, encoding for the A subunit of factor (F)XIII, a transglutaminase that cross-links fibrin and induces clot stabilization. FXIII-A is synthesized by hematopoietic cells, megakaryocytes, and monocytes. Objectives To understand RUNX1 regulation of F13A1 expression in platelets/megakaryocytes and the mechanisms and consequences of decreased F13A1 in RHD. Methods We performed studies in platelets, human erythroleukemia (HEL) cells, and human CD34+ cell-derived megakaryocytes including on clot contraction in cells following small inhibitor RNA knockdown (KD) of RUNX1 or F13A1. Results Platelet F13A1 mRNA and protein were decreased in our index patient and in 2 siblings from an unrelated family with RHD. Platelet-driven clot contraction was decreased in the patient and affected daughter. Promoter studies in HEL cells showed that RUNX1 regulates F13A1 transcription; RUNX1 overexpression increased, and small inhibitor RNA RUNX1 KD reduced F13A1 promoter activity and protein. Following RUNX1 or F13A1 KD, clot contraction by HEL cells was decreased, as were FXIII-A surface expression, myosin light chain phosphorylation, and PAC1 antibody binding upon activation. F13A1 expression and clot contraction were impaired in RUNX1 downregulation in human megakaryocytes. Conclusion RUNX1 regulates platelet-megakaryocyte F13A1 expression, which is decreased in RHD, reflecting regulation of a coagulation protein by a hematopoietic transcription factor. Platelet and megakaryocyte clot contraction is decreased in RHD, related to multiple impaired mechanisms including F13A1 expression, myosin phosphorylation, and αIIbβ3 activation.
Collapse
Affiliation(s)
- Fabiola Del Carpio-Cano
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Natthapol Songdej
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
- Penn State College of Medicine/Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Liying Guan
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Guangfen Mao
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Lawrence E. Goldfinger
- Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jeremy G.T. Wurtzel
- Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Kiwon Lee
- Department of Pediatrics, Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Bioscience and Biotechnology, Hankuk University of Foreign Studies, Seoul, South Korea
| | - Michele P. Lambert
- Department of Pediatrics, Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Mortimer Poncz
- Department of Pediatrics, Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - A. Koneti Rao
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
- Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
10
|
Del Carpio-Cano F, Songdej N, Guan L, Mao G, Goldfinger LE, Wurtzel JGT, Lee K, Lambert MP, Poncz M, Koneti Rao A. Transcription Factor RUNX1 Regulates Coagulation Factor XIII-A ( F13A1 ): Decreased Platelet-Megakaryocyte F13A1 Expression and Clot Contraction in RUNX1 Haplodeficiency. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.17.24318561. [PMID: 39763522 PMCID: PMC11702714 DOI: 10.1101/2024.12.17.24318561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Background Germline RUNX1 haplodeficiency (RHD) is associated with thrombocytopenia, platelet dysfunction and predisposition to myeloid malignancies. Platelet expression profiling of a RHD patient showed decreased F13A1, encoding for the A subunit of factor XIII, a transglutaminase that cross-links fibrin and induces clot stabilization. FXIII-A is synthesized by hematopoietic cells, megakaryocytes and monocytes. Aims To understand RUNX1 regulation of F13A1 expression in platelet/megakaryocyte and the mechanisms and consequences of decreased F13A1 in RHD. Methods We performed studies in platelets, HEL cells and human CD34+ cell-derived megakaryocytes including on clot contraction in cells following small inhibitor (si)RNA knockdown (KD) of RUNX1 or F13A1 . Results Platelet F13A1 mRNA and protein were decreased in our index patient and in two siblings from an unrelated family with RHD. Platelet-driven clot contraction was decreased in the patient and affected daughter. Promoter studies in HEL cells showed that RUNX1 regulates F13A1 transcription; RUNX1 overexpression increased and (si)RNA RUNX1 KD reduced F13A1 promoter activity and protein. Following RUNX1 or F13A1 KD clot contraction by HEL cells was decreased as were FXIII-A surface expression, myosin light chain phosphorylation and PAC1 binding upon activation. F13A1 expression and clot contraction were impaired on RUNX1 downregulation in human megakaryocytes. Conclusions RUNX1 regulates platelet-megakaryocyte F13A1 expression, which is decreased in RHD, reflecting regulation of a coagulation protein by a hematopoietic transcription factor. Platelet and megakaryocyte clot contraction is decreased in RHD, related to multiple impaired mechanisms including F13A1 expression, myosin phosphorylation and αII b β 3 activation. Scientific category - Platelets and thrombopoiesis. Essentials RUNX1 regulates expression of FXIII-A chain ( F13A1) in megakaryocytes (MK) and platelets. Platelet and MK F13A1 expression and clot contraction are decreased in RUNX1 deficiency. MK clot contraction, myosin phosphorylation and PAC1-binding are impaired in F13A1 deficiency. Defective clot contraction in RHD arises from defects in multiple platelet-MK mechanisms.
Collapse
|
11
|
Bai H, Li Z, Zhang W, Thaxton C, Ohashi Y, Gonzalez L, Kano M, Yatsula B, Hwa J, Dardik A. Early thrombus formation is required for eccentric and heterogeneous neointimal hyperplasia under disturbed flow. J Thromb Haemost 2024; 22:3614-3628. [PMID: 39173878 PMCID: PMC11608155 DOI: 10.1016/j.jtha.2024.07.028] [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: 05/28/2024] [Revised: 07/06/2024] [Accepted: 07/24/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Anticoagulation and antiplatelet therapy effectively inhibit neointimal hyperplasia (NIH) in both arterial and venous systems but not in arteriovenous fistulae (AVF). The main site of AVF failure is the juxta-anastomotic area that is characterized by disturbed flow compared with laminar flow in the arterial inflow and the venous outflow. OBJECTIVES We hypothesized that early thrombus formation is required for eccentric and heterogeneous NIH in the presence of disturbed flow. METHODS Needle puncture and sutured AVF were created in C57BL/6 mice, in PF4-Cre × mT/mG reporter mice, and in Wistar rats. Human AVF samples were second-stage basilic vein transpositions. The tissues were examined by histology, immunofluorescence, immunohistochemistry, and en face staining. RESULTS In the presence of disturbed flow, both mouse and human AVF showed eccentric and heterogeneous NIH. Maladapted vein wall was characterized by eccentric and heterogeneous neointima that was composed of a different abundance of thrombus and smooth muscle cells. PF4-cre × mT/mG reporter mice AVF showed that green fluorescent protein-labeled platelets deposit on the wall directly facing the fistula exit with endothelial cell loss and continue to accumulate in the presence of disturbed flow. Neither disturbed flow with limited endothelial cell loss nor nondisturbed flow induced heterogeneous neointima in different animal models. CONCLUSION Early thrombus contributes to late heterogeneous NIH in the presence of disturbed flow. Disturbed flow, large area of endothelial cell loss, and thrombus formation are critical to form eccentric and heterogeneous NIH. Categorization of adapted or maladapted walls may be helpful for therapy targeting heterogeneous NIH.
Collapse
Affiliation(s)
- Hualong Bai
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Zhuo Li
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Weichang Zhang
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Carly Thaxton
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Yuichi Ohashi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Masaki Kano
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA; Department of Cellular and Molecular Physiology, Yale University, New Haven, Connecticut, USA; Department of Surgery, Veteran Affairs Connecticut Healthcare Systems, West Haven, Connecticut, USA.
| |
Collapse
|
12
|
Filatov NS, Khismatullin RR, Bilyalov AI, Khabirova AI, Salyakhutdinova SM, Ursan RV, Kasimova RN, Peshkova AD, Gazizov II, Shagimardanova EI, Woroncow MV, Kiyasov AP, Litvinov RI, Gusev OA. Distinct Hemostasis and Blood Composition in Spiny Mouse Acomys cahirinus. Int J Mol Sci 2024; 25:12867. [PMID: 39684578 DOI: 10.3390/ijms252312867] [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: 10/17/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
The spiny mouse (Acomys species) is capable of scarless wound regeneration through largely yet unknown mechanisms. To investigate whether this capacity is related to peculiarities of the hemostatic system, we studied the blood of Acomys cahirinus in comparison to Mus musculus (Balb/c) to reveal differences in blood composition and clotting in both males and females. In response to surgical manipulations, blood clots formed in wounds of Acomys comprised a stronger hemostatic seal with reduced surgical bleeding in comparison with Balb/c. Acomys demonstrated notably shorter tail bleeding times and elevated clottable fibrinogen levels. Histological analysis revealed that clots from Acomys blood had densely packed fibrin-rich clots with pronounced fibrin segregation from erythrocytes. Acomys exhibited superior plasma clot stiffness as revealed with thromboelastography. The latter two characteristics are likely due to hyperfibrinogenemia. Light transmission platelet aggregometry demonstrated that ADP-induced platelet aggregates in Acomys males are stable, unlike the aggregates formed in the plasma of Balb/c undergoing progressive disaggregation over time. There were no apparent distinctions in platelet contractility and baseline expression of phosphatidylserine. Hematological profiling revealed a reduced erythrocytes count but increased mean corpuscular volume and hemoglobin content in Acomys. These results demonstrate the distinctive hemostatic potential of Acomys cahirinus, which may contribute to their remarkable regenerative capacity.
Collapse
Affiliation(s)
- Nikita S Filatov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Rafael R Khismatullin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Airat I Bilyalov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Loginov Moscow Clinical Scientific Center, 111123 Moscow, Russia
| | - Alina I Khabirova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | | | - Roman V Ursan
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | | | - Alina D Peshkova
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Insaf I Gazizov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Elena I Shagimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Loginov Moscow Clinical Scientific Center, 111123 Moscow, Russia
| | - Mary V Woroncow
- Institute for Regenerative Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Andrey P Kiyasov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Rustem I Litvinov
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Oleg A Gusev
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- LIFT, Life Improvement by Future Technologies Institute, 121205 Moscow, Russia
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| |
Collapse
|
13
|
Zhang S, Jiang T, Li M, Sun H, Wu H, Wu W, Li Y, Jiang H. Graphene-Based Wound Dressings for Wound Healing: Mechanism, Technical Analysis, and Application Status. ACS Biomater Sci Eng 2024; 10:6790-6813. [PMID: 39467733 DOI: 10.1021/acsbiomaterials.4c01142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
The development of novel wound dressings is critical in medical care. Graphene and its derivatives possess excellent biomedical properties, making them highly suitable for various applications in medical dressings. This review provides a comprehensive technical analysis and the current application status of graphene-based medical dressings. Initially, we discuss the chemical structure and the fabrication method of graphene and its derivatives. We then provide a detailed summary of the mechanisms by which graphene materials promote wound repair across the four stages of wound healing. Subsequently, we categorize the types of graphene-based wound dressings and analyze corresponding characteristics. Finally, we analyze the challenges encountered at present and propose solutions regarding future development trends. This paper aims to serve as a reference for further research in skin tissue engineering and the development of innovative graphene-based medical dressings.
Collapse
Affiliation(s)
- Shanguo Zhang
- School of Mechatronics Engineering, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin 150001, People's Republic of China
| | - Tianyi Jiang
- School of Mechatronics Engineering, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin 150001, People's Republic of China
| | - Ming Li
- School of Mechatronics Engineering, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin 150001, People's Republic of China
| | - Haoxiu Sun
- School of Life Sciences, Harbin Institute of Technology, No. 2 Yikuang Street, Harbin 150001, People's Republic of China
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, No. 157 Health Road, Harbin 150001, People's Republic of China
| | - Hao Wu
- School of Mechatronics Engineering, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin 150001, People's Republic of China
| | - Wenlong Wu
- School of Mechatronics Engineering, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin 150001, People's Republic of China
| | - Yu Li
- School of Life Sciences, Harbin Institute of Technology, No. 2 Yikuang Street, Harbin 150001, People's Republic of China
| | - Hongyuan Jiang
- School of Mechatronics Engineering, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin 150001, People's Republic of China
| |
Collapse
|
14
|
Javaid A, KA A, PM S, Arora K, Mudavath SL. Innovative Approaches and Future Directions in the Management and Understanding of Varicose Veins: A Systematic Review. ACS Pharmacol Transl Sci 2024; 7:2971-2986. [PMID: 39421653 PMCID: PMC11480891 DOI: 10.1021/acsptsci.4c00430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 10/19/2024]
Abstract
Varicose veins, a prevalent condition that primarily affects the lower limbs, present significant hurdles in diagnosis and treatment due to their diverse causes. This study dives into the complex hormonal, environmental, and molecular elements that influence varicose vein genesis, emphasizing the need for precise diagnostic methods and changing therapy approaches to improve patient outcomes. It investigates the epidemiology and demographic distribution of varicose veins, delves into their pathophysiology, and assesses diagnostic methods such as duplex ultrasonography and the CEAP classification system. In addition, the study discusses novel therapies such as sclerotherapy and endovenous thermal ablation, as well as the effectiveness of existing diagnostic methods in detecting chronic venous illnesses. By investigating venous wall remodeling and inflammatory pathways, it gives a thorough knowledge of varicose vein formation. The study calls for future research that focuses on patient-centered methods, bioengineering advances, digital health applications, and genetic and molecular studies to improve the accuracy and effectiveness of vascular therapy. As a result, a multidisciplinary literature analysis was done, drawing on insights from vascular medicine, epidemiology, genetics, and pharmacology, to consolidate existing knowledge and identify possibilities to enhance varicose vein diagnosis, treatment, and patient care outcomes.
Collapse
Affiliation(s)
- Aaqib Javaid
- Infectious
Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali, Punjab 140306, India
| | - Abutwaibe KA
- Infectious
Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali, Punjab 140306, India
| | - Sherilraj PM
- Infectious
Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali, Punjab 140306, India
| | - Kanika Arora
- Infectious
Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali, Punjab 140306, India
| | - Shyam Lal Mudavath
- Department
of Animal Biology, School of Life Sciences, University of Hyderabad, Prof. C.R. Rao Road, Gachibowli Hyderabad, Telangana 500046, India
| |
Collapse
|
15
|
Muttiah B, Ng SL, Lokanathan Y, Ng MH, Law JX. Beyond Blood Clotting: The Many Roles of Platelet-Derived Extracellular Vesicles. Biomedicines 2024; 12:1850. [PMID: 39200314 PMCID: PMC11351396 DOI: 10.3390/biomedicines12081850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/23/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Platelet-derived extracellular vesicles (pEVs) are emerging as pivotal players in numerous physiological and pathological processes, extending beyond their traditional roles in hemostasis and thrombosis. As one of the most abundant vesicle types in human blood, pEVs transport a diverse array of bioactive molecules, including growth factors, cytokines, and clotting factors, facilitating crucial intercellular communication, immune regulation, and tissue healing. The unique ability of pEVs to traverse tissue barriers and their biocompatibility position them as promising candidates for targeted drug delivery and regenerative medicine applications. Recent studies have underscored their involvement in cancer progression, viral infections, wound healing, osteoarthritis, sepsis, cardiovascular diseases, rheumatoid arthritis, and atherothrombosis. For instance, pEVs promote tumor progression and metastasis, enhance tissue repair, and contribute to thrombo-inflammation in diseases such as COVID-19. Despite their potential, challenges remain, including the need for standardized isolation techniques and a comprehensive understanding of their mechanisms of action. Current research efforts are focused on leveraging pEVs for innovative anti-cancer treatments, advanced drug delivery systems, regenerative therapies, and as biomarkers for disease diagnosis and monitoring. This review highlights the necessity of overcoming technical hurdles, refining isolation methods, and establishing standardized protocols to fully unlock the therapeutic potential of pEVs. By understanding the diverse functions and applications of pEVs, we can advance their use in clinical settings, ultimately revolutionizing treatment strategies across various medical fields and improving patient outcomes.
Collapse
Affiliation(s)
- Barathan Muttiah
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Sook Luan Ng
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Yogeswaran Lokanathan
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Min Hwei Ng
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| | - Jia Xian Law
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (Y.L.); (M.H.N.)
| |
Collapse
|
16
|
He W, Karmakar A, Kang J, Rowlands G, Schirmacher S, Méndez-Rojano R, Antaki J. In Vitro and In Silico Characterization of the Aggregation of Thrombi on Textured Ventricular Cannula. Ann Biomed Eng 2024; 52:2076-2087. [PMID: 38679660 DOI: 10.1007/s10439-024-03504-1] [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: 11/07/2023] [Accepted: 03/25/2024] [Indexed: 05/01/2024]
Abstract
The unacceptably high stroke rate associated with HeartMate 3 ventricular assist device (VAD) without signs of adherent pump thrombosis is hypothesized to be the result of the emboli produced by the inflow cannula, that are ingested and ejected from the pump. This in vitro and numerical study aimed to emulate the surface features and supraphysiological shear of a ventricular cannula to provide insight into their effect on thrombogenesis. Human whole blood was perfused at calibrated flow rates in a microfluidic channel to achieve shear rates 1000-7500 s-1, comparable to that experienced on the cannula. The channel contained periodic teeth representative of the rough sintered surface of the HeartMate 3 cannula. The deposition of fluorescently labeled platelets was visualized in real time and analyzed with a custom entity tracking algorithm. Numerical simulations of a multi-constituent thrombosis model were performed to simulate laminar blood flow in the channel. The sustained growth of adherent platelets was observed in all shear conditions ( p < 0.05). However, the greatest deposition was observed at the lower shear rates. The location of deposition with respect to the microfluidic teeth was also found to vary with shear rate. This was confirmed by CFD simulation. The entity tracking algorithm revealed the spatial variation of instances of embolic events. This result suggests that the sintered surface of the ventricular cannula may engender unstable thrombi with a greater likelihood of embolization at supraphysiological shear rates.
Collapse
Affiliation(s)
- Wenxuan He
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Abhishek Karmakar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Junhyuk Kang
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Grant Rowlands
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Samuel Schirmacher
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | | | - James Antaki
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
17
|
Stanton K, Philippou H, Ariëns RA. Ischaemic Stroke, Thromboembolism and Clot Structure. Neuroscience 2024; 550:3-10. [PMID: 38453129 DOI: 10.1016/j.neuroscience.2024.02.024] [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: 11/30/2023] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
Ischaemic stroke is a major cause of morbidity and mortality worldwide. Blood clotting and thromboembolism play a central role in the pathogenesis of ischaemic stroke. An increasing number of recent studies indicate changes in blood clot structure and composition in patients with ischaemic stroke. In this review, we aim to summarise and discuss clot structure, function and composition in ischaemic stroke, including its relationships with clinical diagnosis and treatment options such as thrombolysis and thrombectomy. Studies are summarised in which clot structure and composition is analysed both in vitro from patients' plasma samples and ex vivo in thrombi obtained through interventional catheter-mediated thrombectomy. Mechanisms that drive clot composition and architecture such as neutrophil extracellular traps and clot contraction are also discussed. We find that, while in vitro clot structure in plasma samples from ischaemic stroke patients are consistently altered, showing denser clots that are more resistant to fibrinolysis, current data on the composition and architecture of ex vivo clots obtained by thrombectomy are more variable. With the potential of advances in technologies underpinning both the imaging and retrieving of clots, we expect that future studies in this area will generate new data that is of interest for the diagnosis, optimal treatment strategies and clinical management of patients with ischaemic stroke.
Collapse
Affiliation(s)
- Katherine Stanton
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Helen Philippou
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Robert As Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK.
| |
Collapse
|
18
|
Vandelanotte S, De Meyer SF. Acute Ischemic Stroke Thrombus Composition. Neuroscience 2024; 550:11-20. [PMID: 38185279 DOI: 10.1016/j.neuroscience.2023.12.010] [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: 10/27/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/09/2024]
Abstract
Ischemic stroke is caused by a thrombus blocking one or multiple arteries in the brain, resulting in irreversible damage in the associated brain tissue. The aim of therapy is to restore the blood flow as fast as possible. Two recanalization strategies are currently available: pharmacological thrombolysis using recombinant tissue plasminogen activator (rt-PA) and mechanical removal of the thrombus. Despite recent advancements, achieving efficient recanalization remains a challenge. The precise causes of therapy failure are not fully understood but thrombus composition is likely a key factor in successful recanalization. This review explores acute ischemic stroke thrombus composition, its recently identified components, and how it affects stroke treatment. It also discusses how new insights could enhance current recanalization strategies for ischemic stroke patients.
Collapse
Affiliation(s)
| | - Simon F De Meyer
- Laboratory for Thrombosis Research, KU Leuven Kulak, Kortrijk, Belgium.
| |
Collapse
|
19
|
Chernysh IN, Mukhopadhyay S, Johnson TA, Brooks JA, Sarkar R, Weisel JW, Antalis TM. Time-dependent ultrastructural changes during venous thrombogenesis and thrombus resolution. J Thromb Haemost 2024; 22:1675-1688. [PMID: 38492853 PMCID: PMC11139557 DOI: 10.1016/j.jtha.2024.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 02/08/2024] [Accepted: 02/21/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Deep vein thrombosis is a common vascular event that can result in debilitating morbidity and even death due to pulmonary embolism. Clinically, patients with faster resolution of a venous thrombus have improved prognosis, but the detailed structural information regarding changes that occur in a resolving thrombus over time is lacking. OBJECTIVES To define the spatial-morphologic characteristics of venous thrombus formation, propagation, and resolution at the submicron level over time. METHODS Using a murine model of stasis-induced deep vein thrombosis along with scanning electron microscopy and immunohistology, we determine the specific structural, compositional, and morphologic characteristics of venous thrombi formed after 4 days and identify the changes that take place during resolution by day 7. Comparison is made with the structure and composition of venous thrombi formed in mice genetically deficient in plasminogen activator inhibitor type 1. RESULTS As venous thrombus resolution progresses, fibrin exists in different structural forms, and there are dynamic cellular changes in the compositions of leukocytes, platelet aggregates, and red blood cells. Intrathrombus microvesicles are present that are not evident by histology, and red blood cells in the form of polyhedrocytes are an indicator of clot contraction. Structural evidence of fibrinolysis is observed early during thrombogenesis and is accelerated by plasminogen activator inhibitor type 1 deficiency. CONCLUSION The results reveal unique, detailed ultrastructural and compositional insights along with documentation of the dynamic changes that occur during accelerated resolution that are not evident by standard pathologic procedures and can be applied to inform diagnosis and effectiveness of thrombolytic treatments to improve patient outcomes.
Collapse
Affiliation(s)
- Irina N Chernysh
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Subhradip Mukhopadhyay
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Tierra A Johnson
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jacob A Brooks
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Rajabrata Sarkar
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - John W Weisel
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Toni M Antalis
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Research & Development Service, United States Department of Veterans Affairs Maryland Health Care System, Baltimore, Maryland, USA.
| |
Collapse
|
20
|
Sugerman GP, Bechtel GN, Malinowska Z, Parekh SH, Rausch MK. Mechanical properties of clot made from human and bovine whole blood differ significantly. J Mech Behav Biomed Mater 2024; 154:106508. [PMID: 38513312 DOI: 10.1016/j.jmbbm.2024.106508] [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: 08/18/2023] [Revised: 02/24/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
Thromboembolism - that is, clot formation and the subsequent fragmentation of clot - is a leading cause of death worldwide. Clots' mechanical properties are critical determinants of both the embolization process and the pathophysiological consequences thereof. Thus, understanding and quantifying the mechanical properties of clots is important to our ability to treat and prevent thromboembolic disease. However, assessing these properties from in vivo clots is experimentally challenging. Therefore, we and others have turned to studying in vitro clot mimics instead. Unfortunately, there are significant discrepancies in the reported properties of these clot mimics, which have been hypothesized to arise from differences in experimental techniques and blood sources. The goal of our current work is therefore to compare the mechanical behavior of clots made from the two most common sources, human and bovine blood, using the same experimental techniques. To this end, we tested clots under pure shear with and without initial cracks, under cyclic loading, and under stress relaxation. Based on these data, we computed and compared stiffness, strength, work-to-rupture, fracture toughness, relaxation time constants, and prestrain. While clots from both sources behaved qualitatively similarly, they differed quantitatively in almost every metric. We also correlated each mechanical metric to measures of blood composition. Thereby, we traced this inter-species variability in clot mechanics back to significant differences in hematocrit, but not platelet count. Thus, our work suggests that the results of past studies that have used bovine blood to make in vitro mimics - without adjusting blood composition - should be interpreted carefully. Future studies about the mechanical properties of blood clots should focus on human blood alone.
Collapse
Affiliation(s)
- Gabriella P Sugerman
- University of Texas at Austin, Department of Biomedical Engineering, 107 W Dean Keeton St, Austin, TX 78712, United States of America
| | - Grace N Bechtel
- University of Texas at Austin, Department of Biomedical Engineering, 107 W Dean Keeton St, Austin, TX 78712, United States of America
| | - Zuzanna Malinowska
- University of Texas at Austin, Department of Aerospace Engineering & Engineering Mechanics, 2617 Wichita St, Austin, TX 78712, United States of America
| | - Sapun H Parekh
- University of Texas at Austin, Department of Biomedical Engineering, 107 W Dean Keeton St, Austin, TX 78712, United States of America
| | - Manuel K Rausch
- University of Texas at Austin, Department of Biomedical Engineering, 107 W Dean Keeton St, Austin, TX 78712, United States of America; University of Texas at Austin, Department of Aerospace Engineering & Engineering Mechanics, 2617 Wichita St, Austin, TX 78712, United States of America; University of Texas at Austin, Department of Mechanical Engineering, 204 E Dean Keeton St, Austin, TX 78712, United States of America; University of Texas at Austin, Oden Institute for Computational Engineering and Sciences, 201 E 24th St, Austin, TX 78712, United States of America.
| |
Collapse
|
21
|
Wang J, Ho P, Nandurkar H, Lim HY. Overall haemostatic potential assay for prediction of outcomes in venous and arterial thrombosis and thrombo-inflammatory diseases. J Thromb Thrombolysis 2024; 57:852-864. [PMID: 38649560 DOI: 10.1007/s11239-024-02975-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/28/2024] [Indexed: 04/25/2024]
Abstract
Thromboembolic diseases including arterial and venous thrombosis are common causes of morbidity and mortality globally. Thrombosis frequently recurs and can also complicate many inflammatory conditions through the process of 'thrombo-inflammation,' as evidenced during the COVID-19 pandemic. Current candidate biomarkers for thrombosis prediction, such as D-dimer, have poor predictive efficacy. This limits our capacity to tailor anticoagulation duration individually and may expose lower risk individuals to undue bleeding risk. Global coagulation assays, such as the Overall Haemostatic Potential (OHP) assay, that investigate fibrin generation and fibrinolysis, may provide a more accurate and functional assessment of hypercoagulability. We present a review of fibrin's critical role as a central modulator of thrombotic risk. The results of our studies demonstrating the OHP assay as a predictive biomarker in venous thromboembolism, chronic renal disease, diabetes mellitus, post-thrombotic syndrome, and COVID-19 are discussed. As a comprehensive and global measurement of fibrin generation and fibrinolytic capacity, the OHP assay may be a valuable addition to future multi-modal predictive tools in thrombosis.
Collapse
Affiliation(s)
- Julie Wang
- Northern Health, 185 Cooper St, Epping, VIC, 3076, Australia.
| | - Prahlad Ho
- Northern Health, 185 Cooper St, Epping, VIC, 3076, Australia
| | - Harshal Nandurkar
- Australian Centre for Blood Diseases, Monash Health, Melbourne, Australia
| | - Hui Yin Lim
- Northern Health, 185 Cooper St, Epping, VIC, 3076, Australia
| |
Collapse
|
22
|
Peshkova AD, Weisel JW, Litvinov RI. A novel technique to quantify the kinetics of blood clot contraction based on the expulsion of fluorescently labeled albumin into serum. J Thromb Haemost 2024; 22:1742-1748. [PMID: 38401713 PMCID: PMC11139561 DOI: 10.1016/j.jtha.2024.02.012] [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: 01/03/2024] [Revised: 02/09/2024] [Accepted: 02/10/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND The platelet-driven contraction or retraction of blood clots has been utilized to obtain blood serum for laboratory studies, but now, in vitro clot contraction assays are used in research laboratories and clinics to assess platelet functionality. The static final extent of clot contraction measured using a clot size or expelled serum volume can be supplemented substantially with a dynamic analysis. OBJECTIVES To provide a step-by-step protocol for a relatively simple and affordable new automated methodology to follow the kinetics of blood clot contraction, which allows for simultaneous measurements of various samples at a time and requires only a fluorescence plate reader. METHODS The kinetics of clot contraction in whole blood was assessed by continuously detecting the fluorescence intensity of fluorescein isothiocyanate-albumin added to a blood sample before clotting and expelled into the serum during clot shrinkage. RESULTS The clots are formed and fluorescence is measured in the wells of a black multiwell plate using a standard plate fluorescent reader. The specificity of this technique for clot contraction has been demonstrated by the strong inhibitory effects of blebbistatin, latrunculin A, and abciximab. To validate the new technique, increased fluorescence intensity in the contracting clots was measured in parallel with a visual decrease in clot size performed with the same blood samples. CONCLUSION The resulting clot contraction dynamics based on the expulsion of fluorescein isothiocyanate-albumin can be quantified using a number of kinetic parameters as well as a phase kinetics analysis. The advantages and drawbacks of the new technique are discussed.
Collapse
Affiliation(s)
- Alina D Peshkova
- Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - John W Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Rustem I Litvinov
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
23
|
Davila J, O'Brien SH, Mitchell WB, Manwani D. Evaluating thromboprophylaxis in the sickle cell disease population: Navigating the evidence gap. Br J Haematol 2024; 204:2184-2193. [PMID: 38578212 DOI: 10.1111/bjh.19428] [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: 10/02/2023] [Revised: 02/08/2024] [Accepted: 03/14/2024] [Indexed: 04/06/2024]
Abstract
Sickle cell disease (SCD) arises from beta-globin gene mutations, with global estimates indicating around 500 000 affected neonates in 2021. In the United States, it is considered rare, impacting fewer than 200 000 individuals. The key pathogenic flaw lies in mutant haemoglobin S, prone to polymerization under low oxygen conditions, causing erythrocytes to adopt a sickled shape. This leads to complications like vascular occlusion, haemolytic anaemia, inflammation and organ damage. Beyond erythrocyte abnormalities however, there is a body of literature highlighting the hypercoagulable state that is likely a contributor to many of the complications we see in SCD. The persistent activation of the coagulation cascade results in thromboembolic events, notably venous thromboembolism (VTE) which is independently associated with increased mortality in both adults and children with SCD. While the increased risk of VTE in the SCD population seems well established, there is a lack of guidelines for thromboprophylaxis in this population. This Wider Perspective will describe the hypercoagulable state and increased thrombosis risk in the SCD population, as well as advocate for the development of evidence-based guidelines to aid in the prevention of VTE in SCD.
Collapse
Affiliation(s)
- Jennifer Davila
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Albert Einstein College of Medicine, Children's Hospital at Montefiore, Bronx, New York, USA
| | - Sarah H O'Brien
- Division of Pediatric Hematology/Oncology, Nationwide Children's Hospital/The Ohio State University, Columbus, Ohio, USA
| | - William B Mitchell
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Albert Einstein College of Medicine, Children's Hospital at Montefiore, Bronx, New York, USA
| | - Deepa Manwani
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Albert Einstein College of Medicine, Children's Hospital at Montefiore, Bronx, New York, USA
| |
Collapse
|
24
|
Ramanujam RK, Garyfallogiannis K, Litvinov RI, Bassani JL, Weisel JW, Purohit PK, Tutwiler V. Mechanics and microstructure of blood plasma clots in shear driven rupture. SOFT MATTER 2024; 20:4184-4196. [PMID: 38686609 PMCID: PMC11135145 DOI: 10.1039/d4sm00042k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/18/2024] [Indexed: 05/02/2024]
Abstract
Intravascular blood clots are subject to hydrodynamic shear and other forces that cause clot deformation and rupture (embolization). A portion of the ruptured clot can block blood flow in downstream vessels. The mechanical stability of blood clots is determined primarily by the 3D polymeric fibrin network that forms a gel. Previous studies have primarily focused on the rupture of blood plasma clots under tensile loading (Mode I), our current study investigates the rupture of fibrin induced by shear loading (Mode II), dominating under physiological conditions induced by blood flow. Using experimental and theoretical approaches, we show that fracture toughness, i.e. the critical energy release rate, is relatively independent of the type of loading and is therefore a fundamental property of the gel. Ultrastructural studies and finite element simulations demonstrate that cracks propagate perpendicular to the direction of maximum stretch at the crack tip. These observations indicate that locally, the mechanism of rupture is predominantly tensile. Knowledge gained from this study will aid in the development of methods for prediction/prevention of thrombotic embolization.
Collapse
Affiliation(s)
- Ranjini K Ramanujam
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA.
| | | | - Rustem I Litvinov
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - John L Bassani
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia, PA, USA
| | - John W Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Prashant K Purohit
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia, PA, USA
| | - Valerie Tutwiler
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
25
|
Sidonio RF, Weisel JW, Stafford D. Unresolved hemostasis issues in haemophilia. Haemophilia 2024; 30 Suppl 3:70-77. [PMID: 38575518 DOI: 10.1111/hae.14999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 04/06/2024]
Abstract
Despite rapid technological advancement in factor and nonfactor products in the prevention and treatment of bleeding in haemophilia patients, it is imperative that we acknowledge gaps in our understanding of how hemostasis is achieved. The authors will briefly review three unresolved issues in persons with haemophilia (PwH) focusing on the forgotten function that red blood cells play in hemostasis, the critical role of extravascular (outside circulation) FIX in hemostasis in the context of unmodified and extended half-life FIX products and finally on the role that skeletal muscle myosin plays in prothrombinase assembly and subsequent thrombin generation that could mitigate breakthrough muscle hematomas.
Collapse
Affiliation(s)
- Robert F Sidonio
- Hemophilia of Georgia Center for Bleeding and Clotting Disorders of CHOA, Atlanta, Georgia, USA
| | - John W Weisel
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Darrel Stafford
- University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
26
|
Griffin MS, Dahlgren AR, Nagaswami C, Litvinov RI, Keeler K, Madenjian C, Fuentes R, Fish RJ, Neerman-Arbez M, Holinstat M, Adili R, Weisel JW, Shavit JA. Composition of thrombi in zebrafish: similarities and distinctions with mammals. J Thromb Haemost 2024; 22:1056-1068. [PMID: 38160724 PMCID: PMC11293624 DOI: 10.1016/j.jtha.2023.12.025] [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: 08/03/2023] [Revised: 11/28/2023] [Accepted: 12/14/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Blood clots are primarily composed of red blood cells (RBCs), platelets/thrombocytes, and fibrin. Despite the similarities observed between mammals and zebrafish, the composition of fish thrombi is not as well known. OBJECTIVES To analyze the formation of zebrafish blood clots ex vivo and arterial and venous thrombi in vivo. METHODS Transgenic zebrafish lines and laser-mediated endothelial injury were used to determine the relative ratio of RBCs and thrombocytes in clots. Scanning electron and confocal microscopy provided high-resolution images of the structure of adult and larval clots. Adult and larval thrombocyte spreading on fibrinogen was evaluated ex vivo. RESULTS RBCs were present in arterial and venous thrombi, making up the majority of cells in both circulations. However, bloodless mutant fish demonstrated that fibrin clots can form in vivo in the absence of blood cells. Scanning electron and confocal microscopy showed that larval and adult zebrafish thrombi and mammalian thrombi look surprisingly similar externally and internally, even though the former have nucleated RBCs and thrombocytes. Although adult thrombocytes spread on fibrinogen, we found that larval cells do not fully activate without the addition of plasma from adult fish, suggesting a developmental deficiency of a plasma activating factor. Finally, mutants lacking αIIbβ3 demonstrated that this integrin mediates thrombocyte spreading on fibrinogen. CONCLUSION Our data showed strong conservation of arterial and venous and clot/thrombus formation across species, including developmental regulation of thrombocyte function. This correlation supports the possibility that mammals also do not absolutely require circulating cells to form fibrin clots in vivo.
Collapse
Affiliation(s)
- Megan S Griffin
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Anna R Dahlgren
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Chandrasekaran Nagaswami
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Rustem I Litvinov
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kevin Keeler
- US Geological Survey Great Lakes Science Center, Ann Arbor, Michigan, USA
| | - Charles Madenjian
- US Geological Survey Great Lakes Science Center, Ann Arbor, Michigan, USA
| | - Ricardo Fuentes
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Richard J Fish
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Marguerite Neerman-Arbez
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Reheman Adili
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - John W Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jordan A Shavit
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA; Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
27
|
Kim OV, Litvinov RI, Gagne AL, French DL, Brass LF, Weisel JW. Megakaryocyte-induced contraction of plasma clots: cellular mechanisms and structural mechanobiology. Blood 2024; 143:548-560. [PMID: 37944157 PMCID: PMC11033616 DOI: 10.1182/blood.2023021545] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/17/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023] Open
Abstract
ABSTRACT Nonmuscle cell contractility is an essential feature underlying diverse cellular processes such as motility, morphogenesis, division and genome replication, intracellular transport, and secretion. Blood clot contraction is a well-studied process driven by contracting platelets. Megakaryocytes (MKs), which are the precursors to platelets, can be found in bone marrow and lungs. Although they express many of the same proteins and structures found in platelets, little is known about their ability to engage with extracellular proteins such as fibrin and contract. Here, we have measured the ability of MKs to compress plasma clots. Megakaryocytes derived from human induced pluripotent stem cells (iPSCs) were suspended in human platelet-free blood plasma and stimulated with thrombin. Using real-time macroscale optical tracking, confocal microscopy, and biomechanical measurements, we found that activated iPSC-derived MKs (iMKs) caused macroscopic volumetric clot shrinkage, as well as densification and stiffening of the fibrin network via fibrin-attached plasma membrane protrusions undergoing extension-retraction cycles that cause shortening and bending of fibrin fibers. Contraction induced by iMKs involved 2 kinetic phases with distinct rates and durations. It was suppressed by inhibitors of nonmuscle myosin IIA, actin polymerization, and integrin αIIbβ3-fibrin interactions, indicating that the molecular mechanisms of iMK contractility were similar or identical to those in activated platelets. Our findings provide new insights into MK biomechanics and suggest that iMKs can be used as a model system to study platelet contractility. Physiologically, the ability of MKs to contract plasma clots may play a role in the mechanical remodeling of intravascular blood clots and thrombi.
Collapse
Affiliation(s)
- Oleg V. Kim
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Biomedical Engineering and Mechanics, Fralin Biomedical Research Institute, Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA
| | - Rustem I. Litvinov
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Alyssa L. Gagne
- Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Deborah L. French
- Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Lawrence F. Brass
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - John W. Weisel
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW This review highlights how the perception of platelet function is evolving based on recent insights into platelet mechanobiology. RECENT FINDINGS The mechanosensitive ion channel Piezo1 mediates activation of free-flowing platelets under conditions of flow acceleration through mechanisms independent of adhesion receptors and classical activation pathways. Interference with the initiation of platelet migration or with the phenotypic switch of migrating platelets to a procoagulant state aggravates inflammatory bleeding. Mechanosensing of biochemical and biophysical microenvironmental cues during thrombus formation feed into platelet contractile force generation. Measurements of single platelet contraction and bulk clot retraction show promise to identify individuals at risk for hemorrhage. SUMMARY New findings unravel novel mechanotransduction pathways and effector functions in platelets, establishing mechanobiology as a pivotal component of platelet function. These insights highlight limitations of existing treatments and offer new potential therapeutic approaches and diagnostic avenues based on mechanobiological principles. Further extensive research is required to distinguish between core hemostatic and pathological mechanisms influenced by platelet mechanosensing.
Collapse
Affiliation(s)
- Ingmar Schoen
- School of Pharmacy and Biomolecular Sciences
- Irish Centre for Vascular Biology
| | - Martin Kenny
- UCD Conway SPHERE Research Group
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Smita Patil
- School of Pharmacy and Biomolecular Sciences
- Irish Centre for Vascular Biology
| |
Collapse
|
29
|
Lassila R, Weisel JW. Role of red blood cells in clinically relevant bleeding tendencies and complications. J Thromb Haemost 2023; 21:3024-3032. [PMID: 37210074 PMCID: PMC10949759 DOI: 10.1016/j.jtha.2023.05.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/20/2023] [Accepted: 05/10/2023] [Indexed: 05/22/2023]
Abstract
The multiple roles of red blood cells (RBCs) are often neglected as contributors in hemostasis and thrombosis. Proactive opportunities to increase RBC numbers, either acutely or subacutely in the case of iron deficiency, are critical as RBCs are the cellular elements that initiate hemostasis together with platelets and stabilize fibrin and clot structure. RBCs also possess several functional properties to assist hemostasis: releasing platelet agonists, promoting shear force-induced von Willebrand factor unfolding, procoagulant capacity, and binding to fibrin. Additionally, blood clot contraction is important to compress RBCs to form a tightly packed array of polyhedrocytes, making an impermeable seal for hemostasis. All these functions are important for patients having intrinsically poor capacity to cease bleeds (ie, hemostatic disorders) but, conversely, can also play a role in thrombosis if these RBC-mediated reactions overshoot. One acquired example of bleeding with anemia is in patients treated with anticoagulants and/or antithrombotic medication because upon initiation of these drugs, baseline anemia doubles the risk of bleeding complications and mortality. Also, anemia is a risk factor for reoccurring gastrointestinal and urogenital bleeds, pregnancy, and delivery complications. This review summarizes the clinically relevant properties and profiles of RBCs at various steps of platelet adhesion, aggregation, thrombin generation, and fibrin formation, including both structural and functional elements. Regarding patient blood management guidelines, they support minimizing transfusions, but this approach does not deal with severe inherited and acquired bleeding disorders where a poor hemostatic propensity is exacerbated by limited RBC availability, for which future guidance will be needed.
Collapse
Affiliation(s)
- Riitta Lassila
- Research Program Unit in Systems Oncology, Oncosys, Medical Faculty, University of Helsinki, Helsinki, Finland; Coagulation Disorders Unit, Department of Hematology, Helsinki University Hospital, Helsinki, Finland.
| | - John W Weisel
- Department of Cell and Developmental Biology, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
30
|
Zou Y, Shan Z, Han Z, Yang J, Lin Y, Gong Z, Xie L, Xu J, Xie R, Chen Z, Chen Z. Regulating Blood Clot Fibrin Films to Manipulate Biomaterial-Mediated Foreign Body Responses. RESEARCH (WASHINGTON, D.C.) 2023; 6:0225. [PMID: 37719049 PMCID: PMC10503960 DOI: 10.34133/research.0225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/20/2023] [Indexed: 09/19/2023]
Abstract
The clinical efficacy of implanted biomaterials is often compromised by host immune recognition and subsequent foreign body responses (FBRs). During the implantation, biomaterials inevitably come into direct contact with the blood, absorbing blood protein and forming blood clot. Many studies have been carried out to regulate protein adsorption, thus manipulating FBR. However, the role of clot surface fibrin films formed by clotting shrinkage in host reactions and FBR is often ignored. Because of the principle of fibrin film formation being relevant to fibrinogen or clotting factor absorption, it is feasible to manipulate the fibrin film formation via tuning the absorption of fibrinogen and clotting factor. As biological hydroxyapatite reserved bone architecture and microporous structure, the smaller particle size may expose more microporous structures and adsorb more fibrinogen or clotting factor. Therefore, we set up 3 sizes (small, <0.2 mm; medium, 1 to 2 mm; large, 3 to 4 mm) of biological hydroxyapatite (porcine bone-derived hydroxyapatite) with different microporous structures to investigate the absorption of blood protein, the formation of clot surface fibrin films, and the subsequent FBR. We found that small group adsorbed more clotting factors because of more microporous structures and formed the thinnest and sparsest fibrin films. These thinnest and sparsest fibrin films increased inflammation and profibrosis of macrophages through a potential signaling pathway of cell adhesion-cytoskeleton-autophagy, leading to the stronger FBR. Large group adsorbed lesser clotting factors, forming the thickest and densest fibrin films, easing inflammation and profibrosis of macrophages, and finally mitigating FBR. Thus, this study deepens the understanding of the role of fibrin films in host recognition and FBR and demonstrates the feasibility of a strategy to regulate FBR by modulating fibrin films via tuning the absorption of blood proteins.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Zetao Chen
- Hospital of Stomatology, Guanghua School of Stomatology,
Sun Yat-sen University and Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| |
Collapse
|
31
|
Michael C, Pancaldi F, Britton S, Kim OV, Peshkova AD, Vo K, Xu Z, Litvinov RI, Weisel JW, Alber M. Combined computational modeling and experimental study of the biomechanical mechanisms of platelet-driven contraction of fibrin clots. Commun Biol 2023; 6:869. [PMID: 37620422 PMCID: PMC10449797 DOI: 10.1038/s42003-023-05240-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023] Open
Abstract
While blood clot formation has been relatively well studied, little is known about the mechanisms underlying the subsequent structural and mechanical clot remodeling called contraction or retraction. Impairment of the clot contraction process is associated with both life-threatening bleeding and thrombotic conditions, such as ischemic stroke, venous thromboembolism, and others. Recently, blood clot contraction was observed to be hindered in patients with COVID-19. A three-dimensional multiscale computational model is developed and used to quantify biomechanical mechanisms of the kinetics of clot contraction driven by platelet-fibrin pulling interactions. These results provide important biological insights into contraction of platelet filopodia, the mechanically active thin protrusions of the plasma membrane, described previously as performing mostly a sensory function. The biomechanical mechanisms and modeling approach described can potentially apply to studying other systems in which cells are embedded in a filamentous network and exert forces on the extracellular matrix modulated by the substrate stiffness.
Collapse
Affiliation(s)
- Christian Michael
- Department of Mathematics, University of California Riverside, Riverside, CA, 92521, USA
- Center for Quantitative Modeling in Biology, University of California Riverside, Riverside, CA, 92521, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Francesco Pancaldi
- Department of Mathematics, University of California Riverside, Riverside, CA, 92521, USA
- Center for Quantitative Modeling in Biology, University of California Riverside, Riverside, CA, 92521, USA
| | - Samuel Britton
- Department of Mathematics, University of California Riverside, Riverside, CA, 92521, USA
- Center for Quantitative Modeling in Biology, University of California Riverside, Riverside, CA, 92521, USA
| | - Oleg V Kim
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
- Department of Biomedical Engineering and Mechanics, Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Alina D Peshkova
- Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Khoi Vo
- Department of Mathematics, University of California Riverside, Riverside, CA, 92521, USA
- Center for Quantitative Modeling in Biology, University of California Riverside, Riverside, CA, 92521, USA
| | - Zhiliang Xu
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Rustem I Litvinov
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - John W Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA.
| | - Mark Alber
- Department of Mathematics, University of California Riverside, Riverside, CA, 92521, USA.
- Center for Quantitative Modeling in Biology, University of California Riverside, Riverside, CA, 92521, USA.
- Department of Bioengineering, University of California Riverside, Riverside, CA, 92521, USA.
| |
Collapse
|
32
|
Gao D, Sun CW, Woodley AB, Dong JF. Clot Retraction and Its Correlation with the Function of Platelet Integrin α IIbβ 3. Biomedicines 2023; 11:2345. [PMID: 37760786 PMCID: PMC10525596 DOI: 10.3390/biomedicines11092345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 09/29/2023] Open
Abstract
Clot retraction results from retractions of platelet filopodia and fibrin fibers and requires the functional platelet αIIbβ3 integrin. This assay is widely used to test the functions of platelets and fibrinogen as well as the efficacy of fibrinolysis. Changes in clot retraction have been found in a variety of hemostatic abnormalities and, more recently, in arterial thrombosis. Despite its broad clinical use and low cost, many aspects of clot retraction are poorly understood. In the present study, we performed two clinical standard clot retraction assays using whole-blood and platelet-rich plasma (PRP) samples to determine how clot retraction correlates with platelet counts and mean volume, the density of αIIbβ3 integrin and PLA genotypes, and plasma fibrinogen levels. We found that clot retraction was affected by platelet counts, but not mean platelet volume. It correlated with the surface density of the integrin αIibβ3, but not PLA genotypes. These results indicate that clot retraction measures a unique aspect of platelet function and can serve as an additional means to detect functional changes in platelets.
Collapse
Affiliation(s)
- Daniel Gao
- Bloodworks Research Institute, 1551 Southlake Ave. E., Seattle, WA 98102, USA;
- Department of Chemistry, Pomona College, Claremont, CA 91711, USA
| | - Caroline W. Sun
- Section of Cardiovascular Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Angela B. Woodley
- Section of Cardiovascular Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jing-fei Dong
- Bloodworks Research Institute, 1551 Southlake Ave. E., Seattle, WA 98102, USA;
- Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
33
|
Ravera S, Signorello MG, Panfoli I. Platelet Metabolic Flexibility: A Matter of Substrate and Location. Cells 2023; 12:1802. [PMID: 37443836 PMCID: PMC10340290 DOI: 10.3390/cells12131802] [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: 05/17/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Platelets are cellular elements that are physiologically involved in hemostasis, inflammation, thrombotic events, and various human diseases. There is a link between the activation of platelets and their metabolism. Platelets possess considerable metabolic versatility. Although the role of platelets in hemostasis and inflammation is known, our current understanding of platelet metabolism in terms of substrate preference is limited. Platelet activation triggers an oxidative metabolism increase to sustain energy requirements better than aerobic glycolysis alone. In addition, platelets possess extra-mitochondrial oxidative phosphorylation, which could be one of the sources of chemical energy required for platelet activation. This review aims to provide an overview of flexible platelet metabolism, focusing on the role of metabolic compartmentalization in substrate preference, since the metabolic flexibility of stimulated platelets could depend on subcellular localization and functional timing. Thus, developing a detailed understanding of the link between platelet activation and metabolic changes is crucial for improving human health.
Collapse
Affiliation(s)
- Silvia Ravera
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy;
| | | | - Isabella Panfoli
- Department of Pharmacy (DIFAR), University of Genoa, 16132 Genoa, Italy;
| |
Collapse
|