1
|
Jouppila A, Nevola I, Lemponen M, Mattila T, Lassila R. Antiplatelet-anticoagulant, APAC, a mimic of endogenous heparin, is an antithrombotic with von Willebrand factor-mediated characteristics. Thromb Res 2025; 250:109318. [PMID: 40252546 DOI: 10.1016/j.thromres.2025.109318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND We have conjugated selected number of unfractionated heparin (UFH) chains to human albumin core to mimic mast-cell heparin proteoglycans (HEP-PG). Indeed, APAC, dual antiplatelet and anticoagulant, as HEP-PG, has inhibited collagen- (CIPA) and thrombin-induced platelet aggregation, being simultaneously an anticoagulant. In several animal models of arterial thrombosis, APAC has provided vascular-injury-associated local antithrombotic properties mediated by von Willebrand factor (VWF). AIMS We compared the structure-function effects of APAC with those of UFH in vitro, and when supplemented in blood studied platelet and VWF-dependency and anticoagulation. METHODS We assessed the total thrombosis formation analysis system (T-TAS) and coagulation (rotational thromboelastometry, ROTEM) in blood, and thrombin generation and aggregation in platelet-rich plasma. We studied aggregation responses of APAC to collagen, ristocetin, ADP, and potential synergism with cangrelor, P2Y12 receptor antagonist. Finally, heparin-neutralizing role of platelet factor 4 (PF4) on antiplatelet and anticoagulant functions of APAC was investigated. RESULTS APAC concentration-dependently exceeded the anticoagulant and antithrombotic action of UFH in ROTEM, and platelet thrombus formation under arterial blood flow over collagen/tissue factor. APAC uniquely inhibited CIPA. While ADP- and ristocetin-induced aggregation were unaffected by APAC, we detected synergism with cangrelor for CIPA. Disruption of the tertiary structure of APAC reverted its mode of action to anticoagulation only, alike UFH. PF4 neutralized antithrombotic actions of APAC. CONCLUSION The structure-function of APAC conveys dual and unique antiplatelet and anticoagulant actions in flowing blood over collagen and beyond. Our studies confirmed the inhibitory role of APAC on VWF functions and fibrin formation.
Collapse
Affiliation(s)
- Annukka Jouppila
- Clinical Research institute HUCH, Helsinki, Finland; Research Program Unit in Systems Oncology, Medical Faculty, University of Helsinki, Helsinki, Finland.
| | - Ilja Nevola
- Research Program Unit in Systems Oncology, Medical Faculty, University of Helsinki, Helsinki, Finland
| | - Marja Lemponen
- HUS Diagnostic Center, Department of Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tomi Mattila
- Research Program Unit in Systems Oncology, Medical Faculty, University of Helsinki, Helsinki, Finland
| | - Riitta Lassila
- Research Program Unit in Systems Oncology, Medical Faculty, University of Helsinki, Helsinki, Finland; Coagulation Disorders Unit, Departments of Hematology and Clinical Chemistry (HUSLAB Laboratory Services), Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland; Aplagon Ltd., Helsinki, Finland
| |
Collapse
|
2
|
Nakayama M, Goto S, Goto S. Development of the Integrated Computer Simulation Model of the Intracellular, Transmembrane, and Extracellular Domain of Platelet Integrin α IIb β 3 (Platelet Membrane Glycoprotein: GPIIb-IIIa). TH OPEN 2024; 8:e96-e105. [PMID: 38425453 PMCID: PMC10904213 DOI: 10.1055/a-2247-9438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 01/04/2024] [Indexed: 03/02/2024] Open
Abstract
Background The structure and functions of the extracellular domain of platelet integrin α IIb β 3 (platelet membrane glycoprotein: GPIIb-IIIa) change substantially upon platelet activation. However, the stability of the integrated model of extracellular/transmembrane/intracellular domains of integrin α IIb β 3 with the inactive state of the extracellular domain has not been clarified. Methods The integrated model of integrin α IIb β 3 was developed by combining the extracellular domain adopted from the crystal structure and the transmembrane and intracellular domain obtained by Nuclear Magnetic Resonace (NMR). The transmembrane domain was settled into the phosphatidylcholine (2-oleoyl-1-palmitoyl-sn-glycerol-3-phosphocholine (POPC)) lipid bilayer model. The position coordinates and velocity vectors of all atoms and water molecules around them were calculated by molecular dynamic (MD) simulation with the use of Chemistry at Harvard Macromolecular Mechanics force field in every 2 × 10 -15 seconds. Results The root-mean-square deviations (RMSDs) of atoms constructing the integrated α IIb β 3 model apparently stabilized at approximately 23 Å after 200 ns of calculation. However, minor fluctuation persisted during the entire calculation period of 650 ns. The RMSDs of both α IIb and β 3 showed similar trends before 200 ns. The RMSD of β 3 apparently stabilized approximately at 15 Å at 400 ns with persisting minor fluctuation afterward, while the structural fluctuation in α IIb persisted throughout the 650 ns calculation period. Conclusion In conclusion, the integrated model of the intracellular, transmembrane, and extracellular domain of integrin α IIb β 3 suggested persisting fluctuation even after convergence of MD calculation.
Collapse
Affiliation(s)
- Masamitsu Nakayama
- Department of Medicine (Cardiology), Tokai University School of Medicine, Isehara, Japan
| | - Shinichi Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine, Isehara, Japan
| | - Shinya Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
3
|
Hao Y, Závodszky G, Tersteeg C, Barzegari M, Hoekstra AG. Image-based flow simulation of platelet aggregates under different shear rates. PLoS Comput Biol 2023; 19:e1010965. [PMID: 37428797 PMCID: PMC10358939 DOI: 10.1371/journal.pcbi.1010965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/10/2023] [Indexed: 07/12/2023] Open
Abstract
Hemodynamics is crucial for the activation and aggregation of platelets in response to flow-induced shear. In this paper, a novel image-based computational model simulating blood flow through and around platelet aggregates is presented. The microstructure of aggregates was captured by two different modalities of microscopy images of in vitro whole blood perfusion experiments in microfluidic chambers coated with collagen. One set of images captured the geometry of the aggregate outline, while the other employed platelet labelling to infer the internal density. The platelet aggregates were modelled as a porous medium, the permeability of which was calculated with the Kozeny-Carman equation. The computational model was subsequently applied to study hemodynamics inside and around the platelet aggregates. The blood flow velocity, shear stress and kinetic force exerted on the aggregates were investigated and compared under 800 s-1, 1600 s-1 and 4000 s-1 wall shear rates. The advection-diffusion balance of agonist transport inside the platelet aggregates was also evaluated by local Péclet number. The findings show that the transport of agonists is not only affected by the shear rate but also significantly influenced by the microstructure of the aggregates. Moreover, large kinetic forces were found at the transition zone from shell to core of the aggregates, which could contribute to identifying the boundary between the shell and the core. The shear rate and the rate of elongation flow were investigated as well. The results imply that the emerging shapes of aggregates are highly correlated to the shear rate and the rate of elongation. The framework provides a way to incorporate the internal microstructure of the aggregates into the computational model and yields a better understanding of the hemodynamics and physiology of platelet aggregates, hence laying the foundation for predicting aggregation and deformation under different flow conditions.
Collapse
Affiliation(s)
- Yue Hao
- Computational Science Lab, Informatics Institute, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Gábor Závodszky
- Computational Science Lab, Informatics Institute, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
- Department of Hydrodynamic Systems, Budapest University of Technology and Economics, Budapest, Hungary
| | - Claudia Tersteeg
- Laboratory for Thrombosis Research, IRF Life Sciences, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Mojtaba Barzegari
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Alfons G Hoekstra
- Computational Science Lab, Informatics Institute, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Nakayama M, Goto S, Goto S. Physical Characteristics of von Willebrand Factor Binding with Platelet Glycoprotein Ibɑ Mutants at Residue 233 Causing Various Biological Functions. TH OPEN : COMPANION JOURNAL TO THROMBOSIS AND HAEMOSTASIS 2022; 6:e421-e428. [PMID: 36632284 PMCID: PMC9729063 DOI: 10.1055/a-1937-9940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/05/2022] [Indexed: 01/14/2023]
Abstract
Glycoprotein (GP: HIS 1 -PRO 265 ) Ibɑ is a receptor protein expressed on the surface of the platelet. Its N-terminus domain binds with the A1 domain (ASP 1269 -PRO 1472 ) of its ligand protein von Willebrand factor (VWF) and plays a unique role in platelet adhesion under blood flow conditions. Single amino acid substitutions at residue 233 from glycine (G) to alanine (A), aspartic acid (D), or valine (V) are known to cause biochemically distinct functional alterations known as equal, loss, and gain of function, respectively. However, the underlying physical characteristics of VWF binding with GPIbɑ in wild-type and the three mutants exerting different biological functions are unclear. Here, we aimed to test the hypothesis: biological characteristics of macromolecules are influenced by small changes in physical parameters. The position coordinates and velocity vectors of all atoms and water molecules constructing the wild-type and the three mutants of GPIbɑ (G233A, G233D, and G233V) bound with VWF were calculated every 2 × 10 -15 seconds using the CHARMM (Chemistry at Harvard Macromolecular Mechanics) force field for 9 × 10 -10 seconds. Six salt bridges were detected for longer than 50% of the calculation period for the wild-type model generating noncovalent binding energy of -1096 ± 137.6 kcal/mol. In contrast, only four pairs of salt bridges were observed in G233D mutant with noncovalent binding energy of -865 ± 139 kcal/mol. For G233A and G233V, there were six and five pairs of salt bridges generating -929.8 ± 88.5 and -989.9 ± 94.0 kcal/mol of noncovalent binding energy, respectively. Our molecular dynamic simulation showing a lower probability of salt bridge formation with less noncovalent binding energy in VWF binding with the biologically loss of function G233D mutant of GPIbɑ as compared with wild-type, equal function, and gain of function mutant suggests that biological functions of macromolecules such as GPIbɑ are influenced by their small changes in physical characteristics.
Collapse
Affiliation(s)
- Masamitsu Nakayama
- Department of Medicine (Cardiology), Tokai University School of Medicine, Isehara, Japan
| | - Shinichi Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine, Isehara, Japan
| | - Shinya Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine, Isehara, Japan,Address for correspondence Shinya Goto, MD, PhD Department of Medicine (Cardiology), Tokai University School of Medicine143 Shimokasuya, IseharaJapan
| |
Collapse
|
5
|
Tamura N, Goto S, Yokota H, Goto S. Contributing Role of Mitochondrial Energy Metabolism on Platelet Adhesion, Activation and Thrombus Formation under Blood Flow Conditions. Platelets 2022; 33:1083-1089. [PMID: 35348041 DOI: 10.1080/09537104.2022.2046722] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Platelets have an active energy metabolism mediated by mitochondria. However, the role of mitochondria in platelet adhesion, activation, and thrombus formation under blood flow conditions remains to be elucidated. Blood specimens were obtained from healthy adult volunteers. The consumption of glucose molecules by platelets was measured after 24 hours. Platelet adhesion, activation, and thrombus formation on collagen fibrils and immobilized von Willebrand factor (VWF) at a wall shear rate of 1,500 s-1 were detected by fluorescence microscopy with an ultrafast laser confocal unit in the presence or absence of mitochondrial functional inhibitors of carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone (FCCP), antimycin A, and oligomycin. Consumption of glucose molecules within the first 24 h of 4.21 × 10-15 ± 4.46 x 10-15 (n = 6) increased to 13.82 × 10-15 ± 3.46 x 10-15 (n = 4) in the presence of FCCP, 12.11 × 10-15 ± 2.33 x 10-15 (n = 4) in the presence of antimycin A, and 11.87 × 10-15 ± 3.56 x 10-15 (n = 4) in the presence of oligomycin (p < .05). These mitochondrial functional blockers did not influence both surface area coverage by platelets and the 3-dimensional size of platelet thrombi formed on the collagen fibrils. However, a rapid increase in the intracellular calcium ion concentration ([Ca2+]i) upon adhering on immobilized VWF decreased significantly from 405.5 ± 86.2 nM in control to 198.0 ± 79.2 nM in the presence of FCCP (p < .005). A similar decrease in the rapid increase in ([Ca2+]i) was observed in the presence of antimycin A and oligomycin. Mitochondrial function is necessary for platelet activation represented by a rapid increase in [Ca2+]i after platelet adhesion on VWF. However, the influence could not be detected as changes in platelet adhesion or 3-dimensional growth of platelet thrombi on collagen fibrils.
Collapse
Affiliation(s)
- Noriko Tamura
- Department of Health and Nutrition, Faculty of Health Sciences, Niigata University of Health and Welfare, Niigata, Japan
| | - Shinichi Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine, Isehara, Japan
| | - Hideo Yokota
- Image Processing Research Team, Center for Advanced Photonics, Riken, Wako, Japan
| | - Shinya Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
6
|
Structure-Based Cyclic Glycoprotein Ibα-Derived Peptides Interfering with von Willebrand Factor-Binding, Affecting Platelet Aggregation under Shear. Int J Mol Sci 2022; 23:ijms23042046. [PMID: 35216161 PMCID: PMC8876638 DOI: 10.3390/ijms23042046] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 12/25/2022] Open
Abstract
The plasmatic von Willebrand factor (VWF) circulates in a compact form unable to bind platelets. Upon shear stress, the VWF A1 domain is exposed, allowing VWF-binding to platelet glycoprotein Ib-V-IX (GPIbα chain). For a better understanding of the role of this interaction in cardiovascular disease, molecules are needed to specifically interfere with the opened VWF A1 domain interaction with GPIbα. Therefore, we in silico designed and chemically synthetized stable cyclic peptides interfering with the platelet-binding of the VWF A1 domain per se or complexed with botrocetin. Selected peptides (26–34 amino acids) with the lowest-binding free energy were: the monocyclic mono- vOn Willebrand factoR-GPIbα InTerference (ORbIT) peptide and bicyclic bi-ORbIT peptide. Interference of the peptides in the binding of VWF to GPIb-V-IX interaction was retained by flow cytometry in comparison with the blocking of anti-VWF A1 domain antibody CLB-RAg35. In collagen and VWF-dependent whole-blood thrombus formation at a high shear rate, CLB-RAg35 suppressed stable platelet adhesion as well as the formation of multilayered thrombi. Both peptides phenotypically mimicked these changes, although they were less potent than CLB-RAg35. The second-round generation of an improved peptide, namely opt-mono-ORbIT (28 amino acids), showed an increased inhibitory activity under flow. Accordingly, our structure-based design of peptides resulted in physiologically effective peptide-based inhibitors, even for convoluted complexes such as GPIbα-VWF A1.
Collapse
|
7
|
A novel mouse model of type 2N VWD was developed by CRISPR/Cas9 gene editing and recapitulates human type 2N VWD. Blood Adv 2022; 6:2778-2790. [PMID: 35015821 PMCID: PMC9092403 DOI: 10.1182/bloodadvances.2021006353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/21/2021] [Indexed: 11/20/2022] Open
Abstract
A novel type 2N VWD mouse model was established in which VWF is incapable of binding FVIII but is otherwise fully functional. VWF2N/2N mice exhibited a severe bleeding phenotype after tail tip amputation but not in lateral tail vein or ventral artery injury models.
Type 2N von Willebrand disease is caused by mutations in the factor VIII (FVIII) binding site of von Willebrand factor (VWF), resulting in dysfunctional VWF with defective binding capacity for FVIII. We developed a novel type 2N mouse model using CRISPR/Cas9 technology. In homozygous VWF2N/2N mice, plasma VWF levels were normal (1167 ± 257 mU/mL), but the VWF was completely incapable of binding FVIII, resulting in 53 ± 23 mU/mL of plasma FVIII levels that were similar to those in VWF-deficient (VWF−/−) mice. When wild-type human or mouse VWF was infused into VWF2N/2N mice, endogenous plasma FVIII was restored, peaking at 4 to 6 hours post-infusion, demonstrating that FVIII expressed in VWF2N mice is viable but short-lived unprotected in plasma due to dysfunctional 2N VWF. The whole blood clotting time and thrombin generation were impaired in VWF2N/2N but not in VWF−/− mice. Bleeding time and blood loss in VWF2N/2N mice were similar to wild-type mice in the lateral tail vein or ventral artery injury model. However, VWF2N/2N mice, but not VWF−/− mice, lost a significant amount of blood during the primary bleeding phase after a tail tip amputation injury model, indicating that alternative pathways can at least partially restore hemostasis when VWF is absent. In summary, we have developed a novel mouse model by gene editing with both the pathophysiology and clinical phenotype found in severe type 2N patients. This unique model can be used to investigate the biological properties of VWF/FVIII association in hemostasis and beyond.
Collapse
|
8
|
Tamura N, Shimizu K, Shiozaki S, Sugiyama K, Nakayama M, Goto S, Takagi S, Goto S. Important Regulatory Roles of Erythrocytes on Platelet Adhesion to the von Willebrand Factor on the Wall Under Blood Flow Conditions. Thromb Haemost 2021; 122:974-983. [PMID: 34695874 DOI: 10.1055/a-1677-9499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The roles of erythrocytes on platelet adhesion to von Willebrand factor (VWF) on the vessel wall through their membrane glycoprotein (GP)Ibα under blood flow condition is still to be elucidated. Blood specimens containing fluorescently labeled platelet and native, biochemically fixed, or artificial erythrocytes, at various hematocrits were perfused on a surface of VWF immobilized on the wall at a shear rate of 1,500 s-1. Rates of platelet adhesions were measured in each condition. Computer simulation of platelet adhesion to the VWF on the wall at the same shear rate was conducted by solving governing equations with a finite-difference method on K-computer. The rates of platelet adhesion were calculated at various hematocrits conditions in the computational domain of 100 µm (x-axis) x 400 µm (y-axis) x 100 µm (z-axis). Biological experiments demonstrated the positive correlation between the rates of platelet adhesion and hematocrit values in native, fixed, and artificial erythrocytes. (r=0.992, 0.934, and 0.825 respectively, p<0.05 for all). The computer simulation results supported the hematocrit dependent increase in platelet adhesion rates on VWF (94.3/sec at 10%, 185.2/sec at 20%, and 327.9/sec at 30%, respectively). These results suggest the important contributing role of erythrocytes on platelet adhesion to the VWF. The augmented z-axis fluctuation of flowing platelet caused by the physical presence of erythrocytes is speculated as the cause for hematocrit dependent increase in platelet adhesion.
Collapse
Affiliation(s)
- Noriko Tamura
- Niigata University of Health and Welfare, Niigata, Japan
| | - Kazuya Shimizu
- The University of Tokyo Graduate School of Engineering Faculty of Engineering, Bunkyo-ku, Japan
| | - Seiji Shiozaki
- Tokai University School of Medicine Graduate School of Medicine, Isehara, Japan
| | - Kazuyasu Sugiyama
- Osaka University School of Engineering Graduate School of Engineering, Suita, Japan
| | - Masamitsu Nakayama
- Tokai University School of Medicine Graduate School of Medicine, Isehara, Japan
| | - Shinichi Goto
- Department of Cardiology, Keio University School of Medicine Graduate School of Medicine, Shinjuku-ku, Japan
| | - Shu Takagi
- Department of Mechanical Engineering, University of Tokyo, Tokyo, Japan
| | - Shinya Goto
- Department of Medicine, Tokai University, Isehara, Japan
| |
Collapse
|
9
|
A Step toward Combined Platelet and Erythrocyte Recovery. Anesthesiology 2021; 135:200-202. [PMID: 34197571 DOI: 10.1097/aln.0000000000003846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
10
|
MacKeigan DT, Ni T, Shen C, Stratton TW, Ma W, Zhu G, Bhoria P, Ni H. Updated Understanding of Platelets in Thrombosis and Hemostasis: The Roles of Integrin PSI Domains and their Potential as Therapeutic Targets. Cardiovasc Hematol Disord Drug Targets 2021; 20:260-273. [PMID: 33001021 DOI: 10.2174/1871529x20666201001144541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/20/2020] [Accepted: 07/26/2020] [Indexed: 11/22/2022]
Abstract
Platelets are small blood cells known primarily for their ability to adhere and aggregate at injured vessels to arrest bleeding. However, when triggered under pathological conditions, the same adaptive mechanism of platelet adhesion and aggregation may cause thrombosis, a primary cause of heart attack and stroke. Over recent decades, research has made considerable progress in uncovering the intricate and dynamic interactions that regulate these processes. Integrins are heterodimeric cell surface receptors expressed on all metazoan cells that facilitate cell adhesion, movement, and signaling, to drive biological and pathological processes such as thrombosis and hemostasis. Recently, our group discovered that the plexin-semaphorin-integrin (PSI) domains of the integrin β subunits exert endogenous thiol isomerase activity derived from their two highly conserved CXXC active site motifs. Given the importance of redox reactions in integrin activation and its location in the knee region, this PSI domain activity may be critically involved in facilitating the interconversions between integrin conformations. Our monoclonal antibodies against the β3 PSI domain inhibited its thiol isomerase activity and proportionally attenuated fibrinogen binding and platelet aggregation. Notably, these antibodies inhibited thrombosis without significantly impairing hemostasis or causing platelet clearance. In this review, we will update mechanisms of thrombosis and hemostasis, including platelet versatilities and immune-mediated thrombocytopenia, discuss critical contributions of the newly discovered PSI domain thiol isomerase activity, and its potential as a novel target for anti-thrombotic therapies and beyond.
Collapse
Affiliation(s)
- Daniel T MacKeigan
- Department of Physiology, University of Toronto, Toronto, ON M5S, Canada
| | - Tiffany Ni
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Chuanbin Shen
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Tyler W Stratton
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Wenjing Ma
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Guangheng Zhu
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Preeti Bhoria
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Heyu Ni
- Department of Physiology, University of Toronto, Toronto, ON M5S, Canada
| |
Collapse
|
11
|
Liu ZL, Ku DN, Aidun CK. Mechanobiology of shear-induced platelet aggregation leading to occlusive arterial thrombosis: A multiscale in silico analysis. J Biomech 2021; 120:110349. [PMID: 33711601 DOI: 10.1016/j.jbiomech.2021.110349] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022]
Abstract
Occlusive thrombosis in arteries causes heart attacks and strokes. The rapid growth of thrombus at elevated shear rates (~10,000 1/s) relies on shear-induced platelet aggregation (SIPA) thought to come about from the entanglement of von Willebrand factor (VWF) molecules. The mechanism for SIPA is not yet understood in terms of cell- and molecule-level dynamics in fast flowing bloodstreams. Towards this end, we develop a multiscale computational model to recreate SIPA in silico, where the suspension dynamics and interactions of individual platelets and VWF multimers are resolved directly. The platelet-VWF interaction via GP1b-A1 bonds is prescribed with intrinsic binding rates theoretically derived and informed by single-molecule measurements. The model is validated against existing microfluidic SIPA experiments, showing good agreement with the in vitro observations in terms of the morphology, traveling distance and capture time of the platelet aggregates. Particularly, the capture of aggregates can occur in a few milliseconds, comparable to the platelet transit time through pathologic arterial stenotic sections and much shorter than the time for shear-induced platelet activation. The multiscale SIPA simulator provides a cross-scale tool for exploring the biophysical mechanisms of SIPA in silico that are difficult to access with single-molecule measurements or micro-/macro-fluidic assays only.
Collapse
Affiliation(s)
- Zixiang L Liu
- George W. Woodruff School of Mechanical Engineering, and Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, GE 30332, United States.
| | - David N Ku
- George W. Woodruff School of Mechanical Engineering, and Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, GE 30332, United States.
| | - Cyrus K Aidun
- George W. Woodruff School of Mechanical Engineering, and Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, GE 30332, United States.
| |
Collapse
|
12
|
Computer Simulation of Platelet Adhesion around Stent Struts in the Presence and Absence of Tissue Defects around Them. J Interv Cardiol 2021; 2021:8880988. [PMID: 33628146 PMCID: PMC7895599 DOI: 10.1155/2021/8880988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/06/2021] [Accepted: 01/28/2021] [Indexed: 11/17/2022] Open
Abstract
Aim To predict platelet accumulation around stent struts in the presence or absence of tissue defects around them. Methods Computer simulations were performed using virtual platelets implementing the function of the three membrane proteins: glycoprotein (GP) Ibα, GPIIb/IIIa, and GPVI. These platelets were perfused around the stent struts implanted into the vessel wall in the presence or absence of tissue defects around them using within the simulation platform. The number of platelets that adhered around stent struts was calculated by solving the blood flow using Navier-Stokes equation along with the adhesion of membrane protein modeled within the platform. Results Platelet accumulation around stent struts occurred mostly at the downstream region of the stent strut array. The majority of platelets adhered at the downstream of the first bend regardless of the tissue defect status. Platelet adhesion around stent struts occurred more rapidly in the presence of tissue defects. Conclusion Computer simulation using virtual platelets suggested a higher rate of platelet adhesion in the presence of tissue defects around stent struts.
Collapse
|
13
|
Kontos A, Willoughby S, Lushington K, Martin J, Wabnitz D, Dorrian J, Kennedy D. Increased Platelet Aggregation in Children and Adolescents with Sleep-disordered Breathing. Am J Respir Crit Care Med 2020; 202:1560-1566. [PMID: 32628860 DOI: 10.1164/rccm.201911-2229oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Rationale: Sleep-disordered breathing (SDB) is associated with increased vascular resistance in children and adults. Persistent increased vascular resistance damages vascular endothelial cells-a marker of which is increased platelet activation.Objectives: This study compared whole-blood impedance platelet aggregation in children with clinically diagnosed SDB warranting adenotonsillectomy and healthy control subjects.Methods: Thirty children who had SDB warranting intervention clinically diagnosed by experienced pediatric otolaryngologists were recruited from adenotonsillectomy waitlists, and 20 healthy children from the community underwent overnight polysomnography to determine SDB severity (obstructive apnea-hypopnea index). Snoring frequency was collected from parents. In the morning, a fasting blood sample was taken, and whole-blood platelet aggregation was measured.Measurements and Main Results: Children with SDB exhibited increased platelet aggregation to TRAP (thrombin receptor-activating peptide) (children with SDB = 114.8 aggregation units [AU] vs. control subjects = 98.0 AU; P < 0.05) and COL antibody (96.7 vs. 82.2 AU; P < 0.05) and an increased trend in ADP antibody (82.3 vs. 69.2 AU; P < 0.07) but not aspirin dialuminate (82.1 vs. 79.5 AU; P > 0.05). No significant association was observed between either the obstructive apnea-hypopnea index and any aggregation parameter, but parental report of snoring was positively associated with TRAP aggregation (Kendall's τ-c = 0.23; P < 0.05).Conclusions: The finding of increased platelet aggregation is consistent with endothelial damage. This suggests that the profile of cardiovascular changes noted in adults with SDB may also occur in children with SDB.
Collapse
Affiliation(s)
- Anna Kontos
- Department of Respiratory and Sleep Medicine and.,Robinson Research Institute.,Discipline of Paediatrics, School of Medicine, and
| | - Scott Willoughby
- School of Medicine, University of Adelaide, Adelaide, South Australia, Australia; and
| | - Kurt Lushington
- Robinson Research Institute.,Centre for Behaviour, Brain and Body, Justice and Society Unit, University of South Australia, Adelaide, South Australia, Australia
| | - James Martin
- Department of Respiratory and Sleep Medicine and.,Robinson Research Institute.,Discipline of Paediatrics, School of Medicine, and
| | - David Wabnitz
- Department of Otolaryngology, Head and Neck Surgery, Women's and Children's Hospital, Adelaide, South Australia, Australia
| | - Jill Dorrian
- Centre for Behaviour, Brain and Body, Justice and Society Unit, University of South Australia, Adelaide, South Australia, Australia
| | - Declan Kennedy
- Department of Respiratory and Sleep Medicine and.,Robinson Research Institute.,Discipline of Paediatrics, School of Medicine, and
| |
Collapse
|
14
|
Chen Y, Ju LA. Biomechanical thrombosis: the dark side of force and dawn of mechano-medicine. Stroke Vasc Neurol 2020; 5:185-197. [PMID: 32606086 PMCID: PMC7337368 DOI: 10.1136/svn-2019-000302] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 12/19/2022] Open
Abstract
Arterial thrombosis is in part contributed by excessive platelet aggregation, which can lead to blood clotting and subsequent heart attack and stroke. Platelets are sensitive to the haemodynamic environment. Rapid haemodynamcis and disturbed blood flow, which occur in vessels with growing thrombi and atherosclerotic plaques or is caused by medical device implantation and intervention, promotes platelet aggregation and thrombus formation. In such situations, conventional antiplatelet drugs often have suboptimal efficacy and a serious side effect of excessive bleeding. Investigating the mechanisms of platelet biomechanical activation provides insights distinct from the classic views of agonist-stimulated platelet thrombus formation. In this work, we review the recent discoveries underlying haemodynamic force-reinforced platelet binding and mechanosensing primarily mediated by three platelet receptors: glycoprotein Ib (GPIb), glycoprotein IIb/IIIa (GPIIb/IIIa) and glycoprotein VI (GPVI), and their implications for development of antithrombotic 'mechano-medicine' .
Collapse
Affiliation(s)
- Yunfeng Chen
- Molecular Medicine, Scripps Research Institute, La Jolla, California, USA
| | - Lining Arnold Ju
- School of Biomedical Engineering, Heart Research Institute and Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
15
|
Abstract
Von Willebrand factor (VWF) and coagulation factor VIII (FVIII) circulate as a complex in plasma and have a major role in the hemostatic system. VWF has a dual role in hemostasis. It promotes platelet adhesion by anchoring the platelets to the subendothelial matrix of damaged vessels and it protects FVIII from proteolytic degradation. Moreover, VWF is an acute phase protein that has multiple roles in vascular inflammation and is massively secreted from Weibel-Palade bodies upon endothelial cell activation. Activated FVIII on the other hand, together with coagulation factor IX forms the tenase complex, an essential feature of the propagation phase of coagulation on the surface of activated platelets. VWF deficiency, either quantitative or qualitative, results in von Willebrand disease (VWD), the most common bleeding disorder. The deficiency of FVIII is responsible for Hemophilia A, an X-linked bleeding disorder. Here, we provide an overview on the role of the VWF-FVIII interaction in vascular physiology.
Collapse
Affiliation(s)
- Klytaimnistra Kiouptsi
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstrasse 1, Building 708, 55131, Mainz, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Langenbeckstrasse 1, Building 708, 55131, Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Mainz, Germany.
| |
Collapse
|
16
|
Hanson SR, Tucker EI, Latour RA. Blood Coagulation and Blood–Material Interactions. Biomater Sci 2020. [DOI: 10.1016/b978-0-12-816137-1.00052-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
17
|
Ye T, Shi H, Phan-Thien N, Lim CT. The key events of thrombus formation: platelet adhesion and aggregation. Biomech Model Mechanobiol 2019; 19:943-955. [PMID: 31754949 DOI: 10.1007/s10237-019-01262-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 11/11/2019] [Indexed: 01/20/2023]
Abstract
Thrombus formation is a complex, dynamic and multistep process, involving biochemical reactions, mechanical stimulation, hemodynamics, and so on. In this study, we concentrate on its two crucial steps: (i) platelets adhered to a vessel wall, or simply platelet adhesion, and (ii) platelets clumping and arrested to the adherent platelets, named platelet aggregation. We report the first direct simulation of three modes of platelet adhesion, detachment, rolling adhesion and firm adhesion, as well as the formation, disintegration, arrestment and consolidation of platelet plugs. The results show that the bond dissociation in the detachment mode is mainly attributed to a high probability of rupturing bonds, such that any existing bond can be quickly ruptured and all bonds would be completely broken. In the rolling adhesion, however, it is mainly attributed to the strong traction from the shear flow or erythrocytes, causing that the bonds are ruptured at the trailing edge of the platelet. The erythrocytes play an important role in platelet activities, such as the formation, disintegration, arrestment and consolidation of platelet plugs. They exert an aggregate force on platelets, a repulsion at a near distance but an attraction at a far distance to the platelets. This aggregate force can promote platelets to form a plug and/or bring along a part of a platelet plug causing its disintegration. It also greatly influences the arrestment and consolidation of platelet plugs, together with the adhesive force from the thrombus.
Collapse
Affiliation(s)
- Ting Ye
- School of Mathematics, Jilin University, Qianjin Ave. 2699, Changchun, 130012, China.
| | - Huixin Shi
- School of Mathematics, Jilin University, Qianjin Ave. 2699, Changchun, 130012, China
| | - Nhan Phan-Thien
- Department of Mechanical Engineering, National University of Singapore, Singapore, 117576, Singapore
| | - Chwee Teck Lim
- Department of Mechanical Engineering, National University of Singapore, Singapore, 117576, Singapore
| |
Collapse
|
18
|
Goto S, Oka H, Ayabe K, Yabushita H, Nakayama M, Hasebe T, Yokota H, Takagi S, Sano M, Tomita A, Goto S. Prediction of binding characteristics between von Willebrand factor and platelet glycoprotein Ibα with various mutations by molecular dynamic simulation. Thromb Res 2019; 184:129-135. [PMID: 31739151 DOI: 10.1016/j.thromres.2019.10.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 10/12/2019] [Accepted: 10/21/2019] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Binding of platelet glycoprotein (GP)Ibα with von-Willebrand factor (VWF) exclusively mediates the initial platelet adhesion to injured vessel wall. To understand the mechanism of biomedical functions, we calculated the dynamic fluctuating three-dimensional (3D) structures and dissociation energy for GPIbα with various single amino-acid substitution at G233, which location is known to cause significant changes in platelet adhesive characteristics. MATERIAL AND METHODS Molecular dynamics (MD) simulation was utilized to calculate 3D structures and Potential of Mean Force (PMF) for wild-type VWF bound with wild-type, G233A (equal function), G233V (gain of function), and G233D (loss of function) GPIbα. Simulation was done on water-soluble condition with time-step of 2 × 10-15 s using NAnoscale Molecular Dynamics (NAMD) with Chemistry at HARvard Molecular Mechanics (CHARMM) force field. Initial structure for each mutant was obtained by inducing single amino-acid substitution to the stable water-soluble binding structure of wild-type. RESULTS The most stable structures of wild-type VWF bound to GPIbα in wild-type or any mutant did not differ. However, bond dissociation energy defined as difference of PMF between most stable structure and the structure at 65 Å mass center distances in G233D was 4.32 kcal/mol (19.5%) lower than that of wild-type. Approximately, 2.07 kcal/mol energy was required to dissociate VWF from GPIbα with G233V at mass center distance from 48 to 52 Å, which may explain the apparent "gain of function" in G233V. CONCLUSION The mechanism of substantially different biochemical characteristics of GPIbα with mutations in G233 location was predicted from physical movement of atoms constructing these proteins.
Collapse
Affiliation(s)
- Shinichi Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine, Metabolic Disease Research Center, Tokai University Graduate School of Medicine, Isehara, Japan; Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Hideki Oka
- Department of Medicine (Cardiology), Tokai University School of Medicine, Metabolic Disease Research Center, Tokai University Graduate School of Medicine, Isehara, Japan
| | - Kengo Ayabe
- Department of Medicine (Cardiology), Tokai University School of Medicine, Metabolic Disease Research Center, Tokai University Graduate School of Medicine, Isehara, Japan
| | - Hiroto Yabushita
- Department of Medicine (Cardiology), Tokai University School of Medicine, Metabolic Disease Research Center, Tokai University Graduate School of Medicine, Isehara, Japan
| | - Masamitsu Nakayama
- Department of Medicine (Cardiology), Tokai University School of Medicine, Metabolic Disease Research Center, Tokai University Graduate School of Medicine, Isehara, Japan
| | - Terumitsu Hasebe
- Department of Radiology, Tokai University Hachioji Hospital, Tokai University School of Medicine, Hachioji, Tokyo, Japan
| | - Hideo Yokota
- Image Processing Research Team, Center for Advanced Photonics Extreme Photonics Research, RIKEN, Wako, Japan
| | - Shu Takagi
- Department of Mechanical Engineering, The University of Tokyo, Tokyo, Japan
| | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Aiko Tomita
- Department of Clinical Pharmacology, Tokai University School of Medicine, Isehara, Japan
| | - Shinya Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine, Metabolic Disease Research Center, Tokai University Graduate School of Medicine, Isehara, Japan.
| |
Collapse
|
19
|
Wang Y, Wu YP, Han JJ, Zhang MQ, Yang CX, Jiao P, Tian H, Zhu C, Qin SC, Sun XJ, Zhang HT, Zhao XM. Inhibitory effects of hydrogen on in vitro platelet activation and in vivo prevention of thrombosis formation. Life Sci 2019; 233:116700. [DOI: 10.1016/j.lfs.2019.116700] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/22/2019] [Accepted: 07/25/2019] [Indexed: 12/14/2022]
|
20
|
Humanized GPIbα-von Willebrand factor interaction in the mouse. Blood Adv 2019; 2:2522-2532. [PMID: 30287479 DOI: 10.1182/bloodadvances.2018023507] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 09/04/2018] [Indexed: 01/02/2023] Open
Abstract
The interaction of platelet glycoprotein Ibα (GPIbα) with von Willebrand factor (VWF) initiates hemostasis after vascular injury and also contributes to pathological thrombosis. GPIbα binding to the VWF A1 domain (VWFA1) is a target for antithrombotic intervention, but attempts to develop pharmacologic inhibitors have been hindered by the lack of animal models because of the species specificity of the interaction. To address this problem, we generated a knockin mouse with Vwf exon 28-encoding domains A1 and A2 replaced by the human homolog (VWFh28). VWFh28 mice (M1HA) were crossbred with a transgenic mouse strain expressing human GPIbα on platelets (mGPIbαnull;hGPIbαTg; H1MA) to generate a new strain (H1HA) with humanized GPIbα-VWFA1 binding. Plasma VWF levels in the latter 3 strains were similar to those of wild-type mice (M1MA). Compared with the strains that had homospecific GPIbα-VWF pairing (M1MA and H1HA), M1HA mice of those with heterospecific pairing had a markedly greater prolongation of tail bleeding time and attenuation of thrombogenesis after injury to the carotid artery than H1MA mice. Measurements of GPIbα-VWFA1 binding affinity by surface plasmon resonance agreed with the extent of observed functional defects. Ristocetin-induced platelet aggregation was similar in H1HA mouse and human platelet-rich plasma, and it was comparably inhibited by monoclonal antibody NMC-4, which is known to block human GPIbα-VWFA1 binding, which also inhibited FeCl3-induced mouse carotid artery thrombosis. Thus, the H1HA mouse strain is a fully humanized model of platelet GPIbα-VWFA1 binding that provides mechanistic and pharmacologic information relevant to human hemostatic and thrombotic disorders.
Collapse
|
21
|
Potential different impact of inhibition of thrombin function and thrombin generation rate for the growth of thrombi formed at site of endothelial injury under blood flow condition. Thromb Res 2019; 179:121-127. [PMID: 31129447 DOI: 10.1016/j.thromres.2019.05.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/27/2019] [Accepted: 05/08/2019] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Thrombin inhibitor and anti-Xa are now widely used in clinical practice. However, the difference between thrombin inhibitor and anti-Xa in prevention of thrombosis is still to be elucidated. MATERIALS AND METHODS Computer simulator implementing the function of platelet, coagulation, fibrinolysis and blood flow was developed. The function of thrombin is defined as to activated platelet at the rate of 0.01 s-1 and to produce fibrin at the rate of 0.1 s-1 in control. The effect of thrombin inhibitor was settled to reduce the rate of platelet activation and fibrin generation changed from 10 to 100% as compared to the control. The local thrombin generation rate on activated platelet was settled as 1.0 s-1 as a control. The effect of anti-Xa was settled to reduce to thrombin generation rate on activated platelet from 10% to 100% as compared to the control. The sizes of thrombi formed at site of endothelial injury in the presence and absence of thrombin inhibitor and anti-Xa were compared. RESULTS AND CONCLUSIONS The size of thrombi formed by 30-s perfusion of blood at site of endothelial injury reduced both in the presence of thrombin inhibitor and anti-Xa. There was significant positive relationship between thrombin inhibitor effect and the size of formed thrombi with R value of 0.96. (p < 0.0001) However, the sizes of thrombi were not influence by anti-Xa until it decreased 30% or less as compared to control. There was no significant relationship between anti-Xa effect and the size of formed thrombi. (R = 0.39, p = 0.09) Our results suggest the different dose-dependent effects of thrombin inhibitor and anti-Xa on thrombus formation at least in specific conditions. Computer simulation may help to predict quantitative antithrombotic effects of various antithrombotic agents.
Collapse
|
22
|
Structure of von Willebrand factor A1 on polystyrene determined from experimental and calculated sum frequency generation spectra. Biointerphases 2018; 13:06E411. [PMID: 30551688 DOI: 10.1116/1.5056219] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The blood-clotting protein von Willebrand factor (vWF) can be activated by small molecules, high shear stress, and interactions with interfaces. It subsequently binds platelet receptor glycoprotein Ibα (GPIbα) at the surface of platelets, thereby playing a crucial role in blood clotting due to platelet activation, which is an important process to consider in the design of cardiovascular implants and biomaterials used in blood-contacting applications. The influence of surfaces on the activation and the molecular-level structure of surface-bound vWF is largely unknown. Recent studies have indicated that when bound to hydrophobic polystyrene (PS), the A1 domain of vWF remains accessible for GPIbα binding. However, the detailed secondary structure and exact orientation of vWF A1 at the PS surface is still unresolved. Here, the authors resolve these features by studying the system with sum-frequency generation (SFG) spectroscopy. The data are consistent with a scenario where vWF A1 maintains a native secondary structure when bound to PS. Comparison of experimental and calculated SFG spectra combined with previously reported time-of-flight secondary ion mass spectrometry data suggests that A1 assumes an orientation with the GPIbα binding domain oriented away from the solid surface and exposed to the solution phase. This structural information will benefit future in vitro experiments with surface-adsorbed A1 domain and may have relevance for the design of novel blood-contacting biomaterials and wound-healing applications.
Collapse
|
23
|
A shear-dependent NO-cGMP-cGKI cascade in platelets acts as an auto-regulatory brake of thrombosis. Nat Commun 2018; 9:4301. [PMID: 30327468 PMCID: PMC6191445 DOI: 10.1038/s41467-018-06638-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 09/18/2018] [Indexed: 12/31/2022] Open
Abstract
Mechanisms that limit thrombosis are poorly defined. One of the few known endogenous platelet inhibitors is nitric oxide (NO). NO activates NO sensitive guanylyl cyclase (NO-GC) in platelets, resulting in an increase of cyclic guanosine monophosphate (cGMP). Here we show, using cGMP sensor mice to study spatiotemporal dynamics of platelet cGMP, that NO-induced cGMP production in pre-activated platelets is strongly shear-dependent. We delineate a new mode of platelet-inhibitory mechanotransduction via shear-activated NO-GC followed by cGMP synthesis, activation of cGMP-dependent protein kinase I (cGKI), and suppression of Ca2+ signaling. Correlative profiling of cGMP dynamics and thrombus formation in vivo indicates that high cGMP concentrations in shear-exposed platelets at the thrombus periphery limit thrombosis, primarily through facilitation of thrombus dissolution. We propose that an increase in shear stress during thrombus growth activates the NO-cGMP-cGKI pathway, which acts as an auto-regulatory brake to prevent vessel occlusion, while preserving wound closure under low shear. Nitric oxide (NO) inhibits thrombosis in part by stimulating cyclic guanosine monophosphate (cGMP) production and cGMP-dependent protein kinase I (cGKI) activity in platelets. Here, Wen et al. develop a cGMP sensor mouse to follow cGMP dynamics in platelets, and find that shear stress activates NO-cGMP-cGKI signaling during platelet aggregation to limit thrombosis.
Collapse
|
24
|
Chetcuti Zammit S, Koulaouzidis A, Sanders DS, McAlindon ME, Rondonotti E, Yung DE, Sidhu R. Overview of small bowel angioectasias: clinical presentation and treatment options. Expert Rev Gastroenterol Hepatol 2018; 12:125-139. [PMID: 28994309 DOI: 10.1080/17474124.2018.1390429] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Elderly patients with multiple co-morbidities are at an increased risk of developing small bowel angioectasias. Treating these lesions can be both challenging and costly with patients requiring extensive investigations and recurrent admissions for iron infusions and blood transfusions as well as invasive procedures. This review presents treatment options and describes in detail drugs that should be considered whilst taking into account their effectiveness and their safety profile. Areas covered: A PubMed search was carried out using the following keywords: small bowel angiodysplasias, small bowel angioectasias, small bowel bleeding and obscure gastrointestinal bleeding to assess existing evidence. The pathophysiology and risk factors are covered in this review together with appropriate methods of investigation and management. Treatment options discussed are endoscopic measures, surgical options and pharmacotherapy. The role of serum biomarkers is also discussed. Expert commentary: Future work should be directed at alternative drugs with a good safety profile that target biomarkers. Novel pharmacotherapy directed at biomarkers could potentially provide a non-invasive treatment option for angioectasias particularly in the elderly where management can be challenging.
Collapse
Affiliation(s)
| | | | - David S Sanders
- a Gastroenterology Department , Royal Hallamshire Hospital , Sheffield , UK
| | - Mark E McAlindon
- a Gastroenterology Department , Royal Hallamshire Hospital , Sheffield , UK
| | | | - Diana E Yung
- b Endoscopy Unit , the Royal Infirmary of Edinburgh , Edinburgh , UK
| | - Reena Sidhu
- a Gastroenterology Department , Royal Hallamshire Hospital , Sheffield , UK
| |
Collapse
|
25
|
Patel BB, Diao Y. Multiscale assembly of solution-processed organic electronics: the critical roles of confinement, fluid flow, and interfaces. NANOTECHNOLOGY 2018; 29:044004. [PMID: 29176055 DOI: 10.1088/1361-6528/aa9d7c] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Organic semiconducting small molecules and polymers provide a rich phase space for investigating the fundamentals of molecular and hierarchical assembly. Stemming from weak intermolecular interactions, their assembly sensitively depends on processing conditions, which in turn drastically modulate their electronic properties. Much work has gone into molecular design strategies that maximize intermolecular interactions and encourage close packing. Less understood, however, is the non-equilibrium assembly that occurs during the fabrication process (especially solution coating and printing) which is critical to determining thin film morphology across length scales. This encompasses polymorphism and molecular packing at molecular scale, assembly of π-bonding aggregates at the tens of nanometers scale, and the formation of domains at the micron-millimeter device scale. Here, we discuss three phenomena ubiquitous in solution processing of organic electronic thin films: the confinement effect, fluid flows, and interfacial assembly and the role they play in directing assembly. This review focuses on the mechanistic understanding of how assembly outcomes couple closely to the solution processing environment, supported by salient examples from the recent literature.
Collapse
Affiliation(s)
- Bijal B Patel
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, 600 S. Mathews Ave., Urbana, IL 61801, United States of America
| | | |
Collapse
|
26
|
Wilson SJ, Ismat FA, Wang Z, Cerra M, Narayan H, Raftis J, Gray TJ, Connell S, Garonzik S, Ma X, Yang J, Newby DE. PAR4 (Protease-Activated Receptor 4) Antagonism With BMS-986120 Inhibits Human Ex Vivo Thrombus Formation. Arterioscler Thromb Vasc Biol 2017; 38:448-456. [PMID: 29269513 PMCID: PMC5779320 DOI: 10.1161/atvbaha.117.310104] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/13/2017] [Indexed: 12/15/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— BMS-986120 is a novel first-in-class oral PAR4 (protease-activated receptor 4) antagonist with potent and selective antiplatelet effects. We sought to determine for the first time, the effect of BMS-986120 on human ex vivo thrombus formation. Approach and Results— Forty healthy volunteers completed a phase 1 parallel-group PROBE trial (Prospective Randomized Open-Label Blinded End Point). Ex vivo platelet activation, platelet aggregation, and thrombus formation were measured at 0, 2, and 24 hours after (1) oral BMS-986120 (60 mg) or (2) oral aspirin (600 mg) followed at 18 hours with oral aspirin (600 mg) and oral clopidogrel (600 mg). BMS-986120 demonstrated highly selective and reversible inhibition of PAR4 agonist peptide (100 μM)-stimulated P-selectin expression, platelet-monocyte aggregates, and platelet aggregation (P<0.001 for all). Compared with pretreatment, total thrombus area (μm2/mm) at high shear was reduced by 29.2% (95% confidence interval, 18.3%–38.7%; P<0.001) at 2 hours and by 21.4% (9.3%–32.0%; P=0.002) at 24 hours. Reductions in thrombus formation were driven by a decrease in platelet-rich thrombus deposition: 34.8% (19.3%–47.3%; P<0.001) at 2 hours and 23.3% (5.1%–38.0%; P=0.016) at 24 hours. In contrast to aspirin alone, or in combination with clopidogrel, BMS-986120 had no effect on thrombus formation at low shear (P=nonsignificant). BMS-986120 administration was not associated with an increase in coagulation times or serious adverse events. Conclusions— BMS-986120 is a highly selective and reversible oral PAR4 antagonist that substantially reduces platelet-rich thrombus formation under conditions of high shear stress. Our results suggest PAR4 antagonism has major potential as a therapeutic antiplatelet strategy. Clinical Trial Registration— URL: http://www.clinicaltrials.gov. Unique identifier: NCT02439190.
Collapse
Affiliation(s)
- Simon J Wilson
- From the British Heart Foundation Centre for Cardiovascular Science (S.J.W., D.E.N.), Medical Research Council Centre for Inflammation Research (J.R., S.C.), and Edinburgh College of Medicine (T.J.G.), University of Edinburgh, United Kingdom; Bristol Myers Squibb, Princeton, NJ (F.A.I., Z.W., M.C., S.G., X.M., J.Y.); and Royal Infirmary of Edinburgh, United Kingdom (H.N.).
| | - Fraz A Ismat
- From the British Heart Foundation Centre for Cardiovascular Science (S.J.W., D.E.N.), Medical Research Council Centre for Inflammation Research (J.R., S.C.), and Edinburgh College of Medicine (T.J.G.), University of Edinburgh, United Kingdom; Bristol Myers Squibb, Princeton, NJ (F.A.I., Z.W., M.C., S.G., X.M., J.Y.); and Royal Infirmary of Edinburgh, United Kingdom (H.N.)
| | - Zhaoqing Wang
- From the British Heart Foundation Centre for Cardiovascular Science (S.J.W., D.E.N.), Medical Research Council Centre for Inflammation Research (J.R., S.C.), and Edinburgh College of Medicine (T.J.G.), University of Edinburgh, United Kingdom; Bristol Myers Squibb, Princeton, NJ (F.A.I., Z.W., M.C., S.G., X.M., J.Y.); and Royal Infirmary of Edinburgh, United Kingdom (H.N.)
| | - Michael Cerra
- From the British Heart Foundation Centre for Cardiovascular Science (S.J.W., D.E.N.), Medical Research Council Centre for Inflammation Research (J.R., S.C.), and Edinburgh College of Medicine (T.J.G.), University of Edinburgh, United Kingdom; Bristol Myers Squibb, Princeton, NJ (F.A.I., Z.W., M.C., S.G., X.M., J.Y.); and Royal Infirmary of Edinburgh, United Kingdom (H.N.)
| | - Hafid Narayan
- From the British Heart Foundation Centre for Cardiovascular Science (S.J.W., D.E.N.), Medical Research Council Centre for Inflammation Research (J.R., S.C.), and Edinburgh College of Medicine (T.J.G.), University of Edinburgh, United Kingdom; Bristol Myers Squibb, Princeton, NJ (F.A.I., Z.W., M.C., S.G., X.M., J.Y.); and Royal Infirmary of Edinburgh, United Kingdom (H.N.)
| | - Jennifer Raftis
- From the British Heart Foundation Centre for Cardiovascular Science (S.J.W., D.E.N.), Medical Research Council Centre for Inflammation Research (J.R., S.C.), and Edinburgh College of Medicine (T.J.G.), University of Edinburgh, United Kingdom; Bristol Myers Squibb, Princeton, NJ (F.A.I., Z.W., M.C., S.G., X.M., J.Y.); and Royal Infirmary of Edinburgh, United Kingdom (H.N.)
| | - Timothy J Gray
- From the British Heart Foundation Centre for Cardiovascular Science (S.J.W., D.E.N.), Medical Research Council Centre for Inflammation Research (J.R., S.C.), and Edinburgh College of Medicine (T.J.G.), University of Edinburgh, United Kingdom; Bristol Myers Squibb, Princeton, NJ (F.A.I., Z.W., M.C., S.G., X.M., J.Y.); and Royal Infirmary of Edinburgh, United Kingdom (H.N.)
| | - Shea Connell
- From the British Heart Foundation Centre for Cardiovascular Science (S.J.W., D.E.N.), Medical Research Council Centre for Inflammation Research (J.R., S.C.), and Edinburgh College of Medicine (T.J.G.), University of Edinburgh, United Kingdom; Bristol Myers Squibb, Princeton, NJ (F.A.I., Z.W., M.C., S.G., X.M., J.Y.); and Royal Infirmary of Edinburgh, United Kingdom (H.N.)
| | - Samira Garonzik
- From the British Heart Foundation Centre for Cardiovascular Science (S.J.W., D.E.N.), Medical Research Council Centre for Inflammation Research (J.R., S.C.), and Edinburgh College of Medicine (T.J.G.), University of Edinburgh, United Kingdom; Bristol Myers Squibb, Princeton, NJ (F.A.I., Z.W., M.C., S.G., X.M., J.Y.); and Royal Infirmary of Edinburgh, United Kingdom (H.N.)
| | - Xuewen Ma
- From the British Heart Foundation Centre for Cardiovascular Science (S.J.W., D.E.N.), Medical Research Council Centre for Inflammation Research (J.R., S.C.), and Edinburgh College of Medicine (T.J.G.), University of Edinburgh, United Kingdom; Bristol Myers Squibb, Princeton, NJ (F.A.I., Z.W., M.C., S.G., X.M., J.Y.); and Royal Infirmary of Edinburgh, United Kingdom (H.N.)
| | - Jing Yang
- From the British Heart Foundation Centre for Cardiovascular Science (S.J.W., D.E.N.), Medical Research Council Centre for Inflammation Research (J.R., S.C.), and Edinburgh College of Medicine (T.J.G.), University of Edinburgh, United Kingdom; Bristol Myers Squibb, Princeton, NJ (F.A.I., Z.W., M.C., S.G., X.M., J.Y.); and Royal Infirmary of Edinburgh, United Kingdom (H.N.)
| | - David E Newby
- From the British Heart Foundation Centre for Cardiovascular Science (S.J.W., D.E.N.), Medical Research Council Centre for Inflammation Research (J.R., S.C.), and Edinburgh College of Medicine (T.J.G.), University of Edinburgh, United Kingdom; Bristol Myers Squibb, Princeton, NJ (F.A.I., Z.W., M.C., S.G., X.M., J.Y.); and Royal Infirmary of Edinburgh, United Kingdom (H.N.)
| |
Collapse
|
27
|
Jamasbi J, Ayabe K, Goto S, Nieswandt B, Peter K, Siess W. Platelet receptors as therapeutic targets: Past, present and future. Thromb Haemost 2017; 117:1249-1257. [DOI: 10.1160/th16-12-0911] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 04/08/2017] [Indexed: 01/08/2023]
Abstract
SummaryAnti-platelet drugs reduce arterial thrombosis after plaque rupture and erosion, prevent stent thrombosis and are used to prevent and treat myocardial infarction and ischaemic stroke. Some of them may also be helpful in treating less frequent diseases such as thrombotic thrombocytopenic purpura. The present concise review aims to cover current and future developments of anti-platelet drugs interfering with the interaction of von Willebrand factor (VWF) with glycoprotein (GP) Ibα, and directed against GPVI, GPIIb/IIIa (integrin αIIbβ3), the thrombin receptor PAR-1, and the ADP receptor P2Y12. The high expectations of having novel antiplatelet drugs which selectively inhibit arterial thrombosis without interfering with normal haemostasis could possibly be met in the near future.
Collapse
|
28
|
Tran L, Mottaghy K, Arlt-Körfer S, Waluga C, Behbahani M. An experimental study of shear-dependent human platelet adhesion and underlying protein-binding mechanisms in a cylindrical Couette system. ACTA ACUST UNITED AC 2017; 62:383-392. [PMID: 27718478 DOI: 10.1515/bmt-2015-0034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 08/19/2016] [Indexed: 11/15/2022]
Abstract
Undesirable thrombotic reactions count among the most frequent and serious complications for patients who rely on the use of medical devices. To improve the design of medical devices, it is essential to develop a more precise understanding of platelet reactions. Clinical studies and experiments have shown a strong dependence of platelet deposition behavior on the flow. However, today the influence of hemodynamic parameters such as the shear rate on thrombotic reactions is not well understood. For the study of the shear-dependent mechanisms leading to the activation, adhesion and aggregation of platelets, a Couette flow system was used to investigate thrombocyte behavior with regard to well-defined flow conditions at shear-rate values between γ˙=400 $\dot \gamma = {\rm{400}}$ and 1400 1/s. Results were calculated for physiological temperature. It could be shown that the platelet adhesion density increased with increasing shear rates up to γ˙=800 1/s $\dot \gamma = {\rm{800 1/s}}$ and the adhesion pattern was homogeneous. At γ˙=800 1/s, $\dot \gamma = {\rm{800 1/s}},$ a sudden drop in platelet adhesion density occurred and platelets adhered in filaments. Fluorescence microscopy results of von Willebrand factor (vWF) confirm that a shear rate of γ˙=800 1/s $\dot \gamma = {\rm{800 1/s}}$ represents the threshold where a switch of the platelet-binding mechanism from fibrinogen-mediated to vWF-mediated platelet adhesion takes place.
Collapse
|
29
|
Kontos A, Lushington K, Martin J, Schwarz Q, Green R, Wabnitz D, Xu X, M Sokoya E, Willoughby S, Baumert M, Ferrante A, La Forgia M, Kennedy D. Relationship between Vascular Resistance and Sympathetic Nerve Fiber Density in Arterial Vessels in Children With Sleep Disordered Breathing. J Am Heart Assoc 2017; 6:JAHA.117.006137. [PMID: 28716800 PMCID: PMC5586314 DOI: 10.1161/jaha.117.006137] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Background Sleep disordered breathing in children is associated with increased blood flow velocity and sympathetic overactivity. Sympathetic overactivity results in peripheral vasoconstriction and reduced systemic vascular compliance, which increases blood flow velocity during systole. Augmented blood flow velocity is recognized to promote vascular remodeling. Importantly, increased vascular sympathetic nerve fiber density and innervation in early life plays a key role in the development of early‐onset hypertension in animal models. Examination of sympathetic nerve fiber density of the tonsillar arteries in children undergoing adenotonsillectomy for Sleep disordered breathing will address this question in humans. Methods and Results Thirteen children scheduled for adenotonsillectomy to treat sleep disordered breathing underwent pupillometry, polysomnography, flow‐mediated dilation, resting brachial artery blood flow velocity (velocity time integral), and platelet aggregation. The dorsal lingual artery (tonsil) was stained and immunofluorescence techniques used to determine sympathetic nerve fiber density. Sympathetic nerve fiber density was correlated with increased resting velocity time integral (r=0.63; P<0.05) and a lower Neuronal Pupillary Index (r=−0.71, P<0.01), as well as a slower mean pupillary constriction velocity (mean, r=−0.64; P<0.05). A faster resting velocity time integral was associated with a slower peak pupillary constriction velocity (r=−0.77; P<0.01) and higher platelet aggregation to collagen antigen (r=0.64; P<0.05). Slower mean and peak pupillary constriction velocity were associated with higher platelet aggregation scores (P<0.05; P<0.01, respectively). Conclusions These results indicate that sympathetic activity is associated with change in both the function and structure of systemic vasculature in children with sleep disordered breathing.
Collapse
Affiliation(s)
- Anna Kontos
- Robinson's Research Institute, School of Medicine, Discipline of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, Australia
| | - Kurt Lushington
- School of Psychology, Social Work and Social Policy, University of South Australia, Adelaide, Australia
| | - James Martin
- Robinson's Research Institute, School of Medicine, Discipline of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, Australia.,Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, Adelaide, Australia
| | - Quenten Schwarz
- Neurovascular Research Laboratory, Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Ryan Green
- Department of Information Technology, Engineering and the Environment, University of South Australia, Adelaide, Australia
| | - David Wabnitz
- Department of Otolaryngology-Head and Neck Surgery, Women's and Children's Hospital, Adelaide, Australia
| | - Xiangjun Xu
- Neurovascular Research Laboratory, Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Elke M Sokoya
- Department of Human Physiology, Flinders University, Adelaide, South Australia, Australia
| | - Scott Willoughby
- Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, University of Adelaide and Royal Adelaide Hospital, Adelaide, Australia
| | - Mathias Baumert
- School of Electrical and Electronic Engineering, University of Adelaide, Adelaide, Australia
| | - Antonio Ferrante
- Robinson's Research Institute, School of Medicine, Discipline of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, Australia.,Department of Immunology SA Pathology, Schools of Medicine and Biological Science, University of Adelaide, Adelaide, Australia
| | - Melissa La Forgia
- Department of Medical Imaging, Women's and Children's Hospital, Adelaide, Australia
| | - Declan Kennedy
- Robinson's Research Institute, School of Medicine, Discipline of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, Australia.,Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, Adelaide, Australia
| |
Collapse
|
30
|
Impact of shear stress on Src and focal adhesion kinase phosphorylation in fibrinogen-adherent platelets. Blood Coagul Fibrinolysis 2017; 28:279-285. [DOI: 10.1097/mbc.0000000000000593] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
31
|
Gut microbiota regulate hepatic von Willebrand factor synthesis and arterial thrombus formation via Toll-like receptor-2. Blood 2017; 130:542-553. [PMID: 28572286 DOI: 10.1182/blood-2016-11-754416] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 05/22/2017] [Indexed: 12/23/2022] Open
Abstract
The symbiotic gut microbiota play pivotal roles in host physiology and the development of cardiovascular diseases, but the microbiota-triggered pattern recognition signaling mechanisms that impact thrombosis are poorly defined. In this article, we show that germ-free (GF) and Toll-like receptor-2 (Tlr2)-deficient mice have reduced thrombus growth after carotid artery injury relative to conventionally raised controls. GF Tlr2-/- and wild-type (WT) mice were indistinguishable, but colonization with microbiota restored a significant difference in thrombus growth between the genotypes. We identify reduced plasma levels of von Willebrand factor (VWF) and reduced VWF synthesis, specifically in hepatic endothelial cells, as a critical factor that is regulated by gut microbiota and determines thrombus growth in Tlr2-/- mice. Static platelet aggregate formation on extracellular matrix was similarly reduced in GF WT, Tlr2-/- , and heterozygous Vwf+/- mice that are all characterized by a modest reduction in plasma VWF levels. Defective platelet matrix interaction can be restored by exposure to WT plasma or to purified VWF depending on the VWF integrin binding site. Moreover, administration of VWF rescues defective thrombus growth in Tlr2-/- mice in vivo. These experiments delineate an unexpected pathway in which microbiota-triggered TLR2 signaling alters the synthesis of proadhesive VWF by the liver endothelium and favors platelet integrin-dependent thrombus growth.
Collapse
|
32
|
Kamada H, Imai Y, Nakamura M, Ishikawa T, Yamaguchi T. Shear-induced platelet aggregation and distribution of thrombogenesis at stenotic vessels. Microcirculation 2017; 24. [PMID: 28109051 DOI: 10.1111/micc.12355] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/17/2017] [Indexed: 12/31/2022]
Abstract
OBJECTIVE SIPA, which is mediated by vWF, is a key mechanism in arterial thrombosis under an abnormally high shear rate of blood flow. We investigated the influence of SIPA on thrombogenesis, focusing on alterations in blood flow at stenotic vessels. METHODS We carried out a computer simulation of thrombogenesis in stenotic vessels at three different injury positions (ie, upstream, apex, and downstream of the stenosis) to evaluate the effect of SIPA. RESULTS The results demonstrated that thrombus volume increased downstream of the stenosis. In particular, growth was enhanced significantly as blood flow velocity and severity of stenosis increased. The influence of SIPA was induced by continuous exposure to high shear rate; thus, SIPA had a greater effect from the apex to downstream of the stenosis along the vessel wall. The asymmetry of the impact of SIPA contributed to the distribution of the thrombus. Furthermore, we found that the degree of SIPA was prolonged in a stenotic vessel with a distal injury, whereas it was moderate with thrombus growth in a nonstenosed vessel. This occurred because platelets and vWF that underwent a high shear rate around the apex were transported to the region downstream of the stenosis. CONCLUSIONS These results suggest that thrombus formation downstream of the stenosis is easily affected by SIPA and hemodynamics.
Collapse
Affiliation(s)
- Hiroki Kamada
- Department of Diagnostic Radiology, Tohoku University Hospital, Sendai, Japan
| | - Yohsuke Imai
- School of Engineering, Tohoku University, Sendai, Japan
| | - Masanori Nakamura
- Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| | - Takuji Ishikawa
- Department of Finemechanics, Tohoku University, Sendai, Japan
| | - Takami Yamaguchi
- Department of Biomedical Engineering, Tohoku University, Sendai, Japan
| |
Collapse
|
33
|
A General Shear-Dependent Model for Thrombus Formation. PLoS Comput Biol 2017; 13:e1005291. [PMID: 28095402 PMCID: PMC5240924 DOI: 10.1371/journal.pcbi.1005291] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 12/07/2016] [Indexed: 01/03/2023] Open
Abstract
Modeling the transport, activation, and adhesion of platelets is crucial in predicting thrombus formation and growth following a thrombotic event in normal or pathological conditions. We propose a shear-dependent platelet adhesive model based on the Morse potential that is calibrated by existing invivo and invitro experimental data and can be used over a wide range of flow shear rates ( 100<γ˙<28,000s-1). We introduce an Eulerian-Lagrangian model where hemodynamics is solved on a fixed Eulerian grid, while platelets are tracked using a Lagrangian framework. A force coupling method is introduced for bidirectional coupling of platelet motion with blood flow. Further, we couple the calibrated platelet aggregation model with a tissue-factor/contact pathway coagulation cascade, representing the relevant biology of thrombin generation and the subsequent fibrin deposition. The range of shear rates covered by the proposed model encompass venous and arterial thrombosis, ranging from low-shear-rate conditions in abdominal aortic aneurysms and thoracic aortic dissections to thrombosis in stenotic arteries following plaque rupture, where local shear rates are extremely high. Hemostasis (thrombus formation) is the normal physiological response that prevents significant blood loss after vascular injury. The resulting clots can form under different flow conditions in the veins as well as the arteries. The excessive and undesirable formation of clots (i.e., thrombosis) in our circulatory system may lead to significant morbidity and mortality. Some of these pathologies are deep vein thrombosis and pulmonary embolism and atherothrombosis (thrombosis triggered by plaque rupture) in coronary arteries, to name a few. The process of clot formation and growth at a site on a blood vessel wall involves a number of simultaneous processes including: multiple chemical reactions in the coagulation cascade, species transport and platelet adhesion all of which are strongly influenced by the hydrodynamic forces. Numerical models for blood clotting normally focus on one of the processes under a specific flow condition. Here, we propose a general numerical model that encompass a wide range of hemodynamic conditions in the veins and arteries, with individual platelets and their adhesive dynamics included explicitly in the models. Further, we include the biochemistry of coagulation cascade, which is essential to modeling thrombus formation, and couple that to our platelet aggregation model. The simulation results—tested against three different experiments—demonstrate that the proposed model is effective in capturing the invivo and invitro experimental observations.
Collapse
|
34
|
Goto S, Tamura N, Ayabe K, Kato E, Hasebe T, Takagi S, Kawamura Y, Goto S. A method and preliminary results of in silico computer simulation for the formation of mix thrombi with platelet and fibrin. ACTA ACUST UNITED AC 2017. [DOI: 10.17106/jbr.31.30] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Shinichi Goto
- Department of Cardiology, Keio University School of Medicine
- Department of Medicine (Cardiology), Tokai University School of Medicine
| | - Noriko Tamura
- Department of Medicine (Cardiology), Tokai University School of Medicine
| | - Kengo Ayabe
- Department of Medicine (Cardiology), Tokai University School of Medicine
| | - Eri Kato
- Department of Medicine (Cardiology), Tokai University School of Medicine
| | - Terumitsu Hasebe
- Department of Radiology, Tokai University Hachioji Hospital, Tokai University School of Medicine
| | - Shu Takagi
- Department of Bioengineering, The University of Tokyo
| | - Yota Kawamura
- Department of Medicine (Cardiology), Tokai University School of Medicine
| | - Shinya Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine
| |
Collapse
|
35
|
|
36
|
Zlobina KE, Guria GT. Platelet activation risk index as a prognostic thrombosis indicator. Sci Rep 2016; 6:30508. [PMID: 27461235 PMCID: PMC4962318 DOI: 10.1038/srep30508] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/04/2016] [Indexed: 01/28/2023] Open
Abstract
Platelet activation in blood flow under high, overcritical shear rates is initiated by Von Willebrand factor. Despite the large amount of experimental data that have been obtained, the value of the critical shear rate, above which von Willebrand factor starts to activate platelets, is still controversial. Here, we recommend a theoretical approach to elucidate how the critical blood shear rate is dependent on von Willebrand factor size. We derived a diagram of platelet activation according to the shear rate and von Willebrand factor multimer size. We succeeded in deriving an explicit formula for the dependence of the critical shear rate on von Willebrand factor molecule size. The platelet activation risk index was introduced. This index is dependent on the flow conditions, number of monomers in von Willebrand factor, and platelet sensitivity. Probable medical applications of the platelet activation risk index as a universal prognostic index are discussed.
Collapse
Affiliation(s)
- K E Zlobina
- National Research Center for Hematology, 125167, Novy Zykovsky pr. 4, Moscow, Russia
| | - G Th Guria
- National Research Center for Hematology, 125167, Novy Zykovsky pr. 4, Moscow, Russia.,Moscow Institute of Physics and Technology, 141700, Institututski per. 9, Dolgoprudny, Russia
| |
Collapse
|
37
|
Tronic EH, Yakovenko O, Weidner T, Baio JE, Penkala R, Castner DG, Thomas WE. Differential surface activation of the A1 domain of von Willebrand factor. Biointerphases 2016; 11:029803. [PMID: 26968213 PMCID: PMC4788635 DOI: 10.1116/1.4943618] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 02/25/2016] [Accepted: 02/29/2016] [Indexed: 12/19/2022] Open
Abstract
The clotting protein von Willebrand factor (VWF) binds to platelet receptor glycoprotein Ibα (GPIbα) when VWF is activated by chemicals, high shear stress, or immobilization onto surfaces. Activation of VWF by surface immobilization is an important problem in the failure of cardiovascular implants, but is poorly understood. Here, the authors investigate whether some or all surfaces can activate VWF at least in part by affecting the orientation or conformation of the immobilized GPIbα-binding A1 domain of VWF. Platelets binding to A1 adsorbed onto polystyrene surfaces translocated rapidly at moderate and high flow, but detached at low flow, while platelets binding to A1 adsorbed onto glass or tissue-culture treated polystyrene surfaces translocated slowly, and detached only at high flow. Both x-ray photoelectron spectroscopy and conformation independent antibodies reported comparable A1 amounts on all surfaces. Time-of-flight secondary ion mass spectrometry (ToF-SIMS) and near-edge x-ray absorption fine structure spectra suggested differences in orientation on the three surfaces, but none that could explain the biological data. Instead, ToF-SIMS data and binding of conformation-dependent antibodies were consistent with the stabilization of an alternative more activated conformation of A1 by tissue culture polystyrene and especially glass. These studies demonstrate that different material surfaces differentially affect the conformation of adsorbed A1 domain and its biological activity. This is important when interpreting or designing in vitro experiments with surface-adsorbed A1 domain, and is also of likely relevance for blood-contacting biomaterials.
Collapse
Affiliation(s)
- Elaine H Tronic
- Department of Bioengineering, University of Washington, Seattle, Washington 98195
| | - Olga Yakovenko
- Department of Bioengineering, University of Washington, Seattle, Washington 98195
| | - Tobias Weidner
- Department of Bioengineering, University of Washington, Seattle, Washington 98195
| | - Joe E Baio
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195
| | - Rebecca Penkala
- Department of Bioengineering, University of Washington, Seattle, Washington 98195
| | - David G Castner
- Departments of Bioengineering and Chemical Engineering, University of Washington, Seattle, Washington 98195
| | - Wendy E Thomas
- Department of Bioengineering, University of Washington, Seattle, Washington 98195
| |
Collapse
|
38
|
|
39
|
Brass LF, Diamond SL. Transport physics and biorheology in the setting of hemostasis and thrombosis. J Thromb Haemost 2016; 14:906-17. [PMID: 26848552 PMCID: PMC4870125 DOI: 10.1111/jth.13280] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/20/2016] [Accepted: 01/26/2016] [Indexed: 02/02/2023]
Abstract
The biophysics of blood flow can dictate the function of molecules and cells in the vasculature with consequent effects on hemostasis, thrombosis, embolism, and fibrinolysis. Flow and transport dynamics are distinct for (i) hemostasis vs. thrombosis and (ii) venous vs. arterial episodes. Intraclot transport changes dramatically the moment hemostasis is achieved or the moment a thrombus becomes fully occlusive. With platelet concentrations that are 50- to 200-fold greater than platelet-rich plasma, clots formed under flow have a different composition and structure compared with blood clotted statically in a tube. The platelet-rich, core/shell architecture is a prominent feature of self-limiting hemostatic clots formed under flow. Importantly, a critical threshold concentration of surface tissue factor is required for fibrin generation under flow. Once initiated by wall-derived tissue factor, thrombin generation and its spatial propagation within a clot can be modulated by γ'-fibrinogen incorporated into fibrin, engageability of activated factor (FIXa)/activated FVIIIa tenase within the clot, platelet-derived polyphosphate, transclot permeation, and reduction of porosity via platelet retraction. Fibrin imparts tremendous strength to a thrombus to resist embolism up to wall shear stresses of 2400 dyne cm(-2) . Extreme flows, as found in severe vessel stenosis or in mechanical assist devices, can cause von Willebrand factor self-association into massive fibers along with shear-induced platelet activation. Pathological von Willebrand factor fibers are A Disintegrin And Metalloprotease with ThromboSpondin-1 domain 13 resistant but are a substrate for fibrin generation due to FXIIa capture. Recently, microfluidic technologies have enhanced the ability to interrogate blood in the context of stenotic flows, acquired von Willebrand disease, hemophilia, traumatic bleeding, and drug action.
Collapse
Affiliation(s)
- Lawrence F. Brass
- Departments of Medicine and Systems Pharmacology, University of Pennsylvania, Philadelphia, PA, USA
| | - Scott L. Diamond
- Departments of Medicine and Systems Pharmacology, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Medicine and Engineering, Department of Chemical Engineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
40
|
Vasan SK, Rostgaard K, Majeed A, Ullum H, Titlestad KE, Pedersen OBV, Erikstrup C, Nielsen KR, Melbye M, Nyrén O, Hjalgrim H, Edgren G. ABO Blood Group and Risk of Thromboembolic and Arterial Disease: A Study of 1.5 Million Blood Donors. Circulation 2016; 133:1449-57; discussion 1457. [PMID: 26939588 DOI: 10.1161/circulationaha.115.017563] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 02/24/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND ABO blood groups have been shown to be associated with increased risks of venous thromboembolic and arterial disease. However, the reported magnitude of this association is inconsistent and is based on evidence from small-scale studies. METHODS AND RESULTS We used the SCANDAT2 (Scandinavian Donations and Transfusions) database of blood donors linked with other nationwide health data registers to investigate the association between ABO blood groups and the incidence of first and recurrent venous thromboembolic and arterial events. Blood donors in Denmark and Sweden between 1987 and 2012 were followed up for diagnosis of thromboembolism and arterial events. Poisson regression models were used to estimate incidence rate ratios as measures of relative risk. A total of 9170 venous and 24 653 arterial events occurred in 1 112 072 individuals during 13.6 million person-years of follow-up. Compared with blood group O, non-O blood groups were associated with higher incidence of both venous and arterial thromboembolic events. The highest rate ratios were observed for pregnancy-related venous thromboembolism (incidence rate ratio, 2.22; 95% confidence interval, 1.77-2.79), deep vein thrombosis (incidence rate ratio, 1.92; 95% confidence interval, 1.80-2.05), and pulmonary embolism (incidence rate ratio, 1.80; 95% confidence interval, 1.71-1.88). CONCLUSIONS In this healthy population of blood donors, non-O blood groups explain >30% of venous thromboembolic events. Although ABO blood groups may potentially be used with available prediction systems for identifying at-risk individuals, its clinical utility requires further comparison with other risk markers.
Collapse
Affiliation(s)
- Senthil K Vasan
- From Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (S.K.V., A.M., O.N., G.E.); Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark (K.R., M.M., H.H.); Department of Clinical Immunology, Blood Bank, Rigshospitalet, University Hospital of Copenhagen, Denmark (H.U.); Department of Clinical Immunology, Odense University Hospital, Denmark (K.-E.T.); Department of Clinical Immunology, Næstved Hospital, Denmark (O.B.V.P.); Department of Clinical Immunology, Aarhus University Hospital, Denmark (C.E.); Department of Clinical Immunology, Aalborg University Hospital, Denmark (K.R.N.); and Hematology Centre, Karolinska University Hospital, Stockholm, Sweden (A.M., G.E.).
| | - Klaus Rostgaard
- From Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (S.K.V., A.M., O.N., G.E.); Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark (K.R., M.M., H.H.); Department of Clinical Immunology, Blood Bank, Rigshospitalet, University Hospital of Copenhagen, Denmark (H.U.); Department of Clinical Immunology, Odense University Hospital, Denmark (K.-E.T.); Department of Clinical Immunology, Næstved Hospital, Denmark (O.B.V.P.); Department of Clinical Immunology, Aarhus University Hospital, Denmark (C.E.); Department of Clinical Immunology, Aalborg University Hospital, Denmark (K.R.N.); and Hematology Centre, Karolinska University Hospital, Stockholm, Sweden (A.M., G.E.)
| | - Ammar Majeed
- From Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (S.K.V., A.M., O.N., G.E.); Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark (K.R., M.M., H.H.); Department of Clinical Immunology, Blood Bank, Rigshospitalet, University Hospital of Copenhagen, Denmark (H.U.); Department of Clinical Immunology, Odense University Hospital, Denmark (K.-E.T.); Department of Clinical Immunology, Næstved Hospital, Denmark (O.B.V.P.); Department of Clinical Immunology, Aarhus University Hospital, Denmark (C.E.); Department of Clinical Immunology, Aalborg University Hospital, Denmark (K.R.N.); and Hematology Centre, Karolinska University Hospital, Stockholm, Sweden (A.M., G.E.)
| | - Henrik Ullum
- From Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (S.K.V., A.M., O.N., G.E.); Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark (K.R., M.M., H.H.); Department of Clinical Immunology, Blood Bank, Rigshospitalet, University Hospital of Copenhagen, Denmark (H.U.); Department of Clinical Immunology, Odense University Hospital, Denmark (K.-E.T.); Department of Clinical Immunology, Næstved Hospital, Denmark (O.B.V.P.); Department of Clinical Immunology, Aarhus University Hospital, Denmark (C.E.); Department of Clinical Immunology, Aalborg University Hospital, Denmark (K.R.N.); and Hematology Centre, Karolinska University Hospital, Stockholm, Sweden (A.M., G.E.)
| | - Kjell-Einar Titlestad
- From Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (S.K.V., A.M., O.N., G.E.); Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark (K.R., M.M., H.H.); Department of Clinical Immunology, Blood Bank, Rigshospitalet, University Hospital of Copenhagen, Denmark (H.U.); Department of Clinical Immunology, Odense University Hospital, Denmark (K.-E.T.); Department of Clinical Immunology, Næstved Hospital, Denmark (O.B.V.P.); Department of Clinical Immunology, Aarhus University Hospital, Denmark (C.E.); Department of Clinical Immunology, Aalborg University Hospital, Denmark (K.R.N.); and Hematology Centre, Karolinska University Hospital, Stockholm, Sweden (A.M., G.E.)
| | - Ole B V Pedersen
- From Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (S.K.V., A.M., O.N., G.E.); Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark (K.R., M.M., H.H.); Department of Clinical Immunology, Blood Bank, Rigshospitalet, University Hospital of Copenhagen, Denmark (H.U.); Department of Clinical Immunology, Odense University Hospital, Denmark (K.-E.T.); Department of Clinical Immunology, Næstved Hospital, Denmark (O.B.V.P.); Department of Clinical Immunology, Aarhus University Hospital, Denmark (C.E.); Department of Clinical Immunology, Aalborg University Hospital, Denmark (K.R.N.); and Hematology Centre, Karolinska University Hospital, Stockholm, Sweden (A.M., G.E.)
| | - Christian Erikstrup
- From Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (S.K.V., A.M., O.N., G.E.); Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark (K.R., M.M., H.H.); Department of Clinical Immunology, Blood Bank, Rigshospitalet, University Hospital of Copenhagen, Denmark (H.U.); Department of Clinical Immunology, Odense University Hospital, Denmark (K.-E.T.); Department of Clinical Immunology, Næstved Hospital, Denmark (O.B.V.P.); Department of Clinical Immunology, Aarhus University Hospital, Denmark (C.E.); Department of Clinical Immunology, Aalborg University Hospital, Denmark (K.R.N.); and Hematology Centre, Karolinska University Hospital, Stockholm, Sweden (A.M., G.E.)
| | - Kaspar Rene Nielsen
- From Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (S.K.V., A.M., O.N., G.E.); Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark (K.R., M.M., H.H.); Department of Clinical Immunology, Blood Bank, Rigshospitalet, University Hospital of Copenhagen, Denmark (H.U.); Department of Clinical Immunology, Odense University Hospital, Denmark (K.-E.T.); Department of Clinical Immunology, Næstved Hospital, Denmark (O.B.V.P.); Department of Clinical Immunology, Aarhus University Hospital, Denmark (C.E.); Department of Clinical Immunology, Aalborg University Hospital, Denmark (K.R.N.); and Hematology Centre, Karolinska University Hospital, Stockholm, Sweden (A.M., G.E.)
| | - Mads Melbye
- From Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (S.K.V., A.M., O.N., G.E.); Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark (K.R., M.M., H.H.); Department of Clinical Immunology, Blood Bank, Rigshospitalet, University Hospital of Copenhagen, Denmark (H.U.); Department of Clinical Immunology, Odense University Hospital, Denmark (K.-E.T.); Department of Clinical Immunology, Næstved Hospital, Denmark (O.B.V.P.); Department of Clinical Immunology, Aarhus University Hospital, Denmark (C.E.); Department of Clinical Immunology, Aalborg University Hospital, Denmark (K.R.N.); and Hematology Centre, Karolinska University Hospital, Stockholm, Sweden (A.M., G.E.)
| | - Olof Nyrén
- From Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (S.K.V., A.M., O.N., G.E.); Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark (K.R., M.M., H.H.); Department of Clinical Immunology, Blood Bank, Rigshospitalet, University Hospital of Copenhagen, Denmark (H.U.); Department of Clinical Immunology, Odense University Hospital, Denmark (K.-E.T.); Department of Clinical Immunology, Næstved Hospital, Denmark (O.B.V.P.); Department of Clinical Immunology, Aarhus University Hospital, Denmark (C.E.); Department of Clinical Immunology, Aalborg University Hospital, Denmark (K.R.N.); and Hematology Centre, Karolinska University Hospital, Stockholm, Sweden (A.M., G.E.)
| | - Henrik Hjalgrim
- From Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (S.K.V., A.M., O.N., G.E.); Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark (K.R., M.M., H.H.); Department of Clinical Immunology, Blood Bank, Rigshospitalet, University Hospital of Copenhagen, Denmark (H.U.); Department of Clinical Immunology, Odense University Hospital, Denmark (K.-E.T.); Department of Clinical Immunology, Næstved Hospital, Denmark (O.B.V.P.); Department of Clinical Immunology, Aarhus University Hospital, Denmark (C.E.); Department of Clinical Immunology, Aalborg University Hospital, Denmark (K.R.N.); and Hematology Centre, Karolinska University Hospital, Stockholm, Sweden (A.M., G.E.)
| | - Gustaf Edgren
- From Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden (S.K.V., A.M., O.N., G.E.); Department of Epidemiology Research, Statens Serum Institute, Copenhagen, Denmark (K.R., M.M., H.H.); Department of Clinical Immunology, Blood Bank, Rigshospitalet, University Hospital of Copenhagen, Denmark (H.U.); Department of Clinical Immunology, Odense University Hospital, Denmark (K.-E.T.); Department of Clinical Immunology, Næstved Hospital, Denmark (O.B.V.P.); Department of Clinical Immunology, Aarhus University Hospital, Denmark (C.E.); Department of Clinical Immunology, Aalborg University Hospital, Denmark (K.R.N.); and Hematology Centre, Karolinska University Hospital, Stockholm, Sweden (A.M., G.E.)
| |
Collapse
|
41
|
Zhou F, Cui YY, Wu LL, Yang J, Liu L, Maitz MF, Brown IG, Huang N. Analysis of Flow Field in Mechanical Aortic Bileaflet Heart Valves Using Finite Volume Method. J Med Biol Eng 2016. [DOI: 10.1007/s40846-016-0106-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
42
|
|
43
|
Shiozaki S, Takagi S, Goto S. Prediction of Molecular Interaction between Platelet Glycoprotein Ibα and von Willebrand Factor using Molecular Dynamics Simulations. J Atheroscler Thromb 2015; 23:455-64. [PMID: 26581184 DOI: 10.5551/jat.32458] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM The molecular mechanism of the unique interaction between platelet membrane glycoprotein Ibα (GPIbα) and von Willebrand Factor (VWF), necessary for platelet adhesion under high shear stress, is yet to be clarified. METHODS The molecular dynamics simulation using NAMD (Nanoscale Molecular Dynamics) package with the CHARMM 22 (Chemistry at Harvard Macromolecular Mechanics) force field were used to predict dynamic structural changes occurring in the binding site of A1 domain of VWF and N terminus domain of GPIbα under water soluble condition. RESULTS The mean distance between the mass center of A1 domain of VWF and GPIbα in the stable form was predicted as 27.3 Å. The potential of mean force between the A1 domain of VWF and GPIbα were calculated in conditions of various distances of the mass center between them. All the calculated values were fitted to the Morse potential energy function curve. The maximum adhesive force between A1 domain of VWF and GPIbα was predicted as 62.3 pN by differentiating the potential of mean force with respect to the molecular distance. CONCLUSIONS The molecular dynamics simulation is useful for predicting the dynamic structure changes of protein bonds involved in platelet adhesion and for predicting the adhesive forces generated between their interactions.
Collapse
Affiliation(s)
- Seiji Shiozaki
- Department of Medicine (Cardiology), Tokai University School of Medicine
| | | | | |
Collapse
|
44
|
Tobimatsu H, Nishibuchi Y, Sudo R, Goto S, Tanishita K. Adhesive Forces between A1 Domain of von Willebrand Factor and N-terminus Domain of Glycoprotein Ibα Measured by Atomic Force Microscopy. J Atheroscler Thromb 2015; 22:1091-9. [PMID: 25972025 DOI: 10.5551/jat.28423] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM von Willebrand factor (VWF) plays an important role in the regulation of hemostasis and thrombosis formation, particularly under a high shear rate. However, the adhesive force due to the molecular interaction between VWF and glycoprotein Ibα (GPIbα) has not been fully explored. Thus, we employed atomic force microscopy to directly measure the adhesive force between VWF and GPIbα. METHODS We measured the adhesive force between VWF and GPIbα at the molecular level using an atomic force microscope (AFM). An AFM cantilever was coated with recombinant N-terminus VWF binding site of GPIbα, whereas a cover glass was coated with native VWF. RESULTS The adhesive force at the molecular level was measured using an AFM. In the presence of 1 μg/mL VWF, the adhesion force was nearly 200 pN. As per the Gaussian fit analysis, the adhesive force of a single bond could have been 54 or 107 pN. CONCLUSION Our consideration with the Gaussian fit analysis proposed that the adhesive force of a single bond could be 54 pN, which is very close to that obtained by optical tweezers (50 pN).
Collapse
|
45
|
Harris DG, Benipal PK, Cheng X, Burdorf L, Azimzadeh AM, Pierson RN. Four-dimensional characterization of thrombosis in a live-cell, shear-flow assay: development and application to xenotransplantation. PLoS One 2015; 10:e0123015. [PMID: 25830912 PMCID: PMC4382176 DOI: 10.1371/journal.pone.0123015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 02/26/2015] [Indexed: 02/01/2023] Open
Abstract
Background Porcine xenografts are a promising source of scarce transplantable organs, but stimulate intense thrombosis of human blood despite targeted genetic and pharmacologic interventions. Current experimental models do not enable study of the blood/endothelial interface to investigate adhesive interactions and thrombosis at the cellular level under physiologic conditions. The purpose of this study was to develop and validate a live-cell, shear-flow based thrombosis assay relevant to general thrombosis research, and demonstrate its potential in xenotransplantation applications. Methodology/Principal Findings Confluent wild-type (WT, n = 48) and Gal transferase knock-out (GalTKO, which resist hyperacute rejection; n = 11) porcine endothelia were cultured in microfluidic channels. To mimic microcirculatory flow, channels were perfused at 5 dynes/cm2 and 37°C with human blood stained to fluorescently label platelets. Serial fluorescent imaging visualized percent surface area coverage (SA, for adhesion of labeled cells) and total fluorescence (a metric of clot volume). Aggregation was calculated by the fluorescence/SA ratio (FR). WT endothelia stimulated diffuse platelet adhesion (SA 65 ± 2%) and aggregation (FR 120 ± 1 a.u.), indicating high-grade thrombosis consistent with the rapid platelet activation and consumption seen in whole-organ lung xenotransplantation models. Experiments with antibody blockade of platelet aggregation, and perfusion of syngeneic and allo-incompatible endothelium was used to verify the biologic specificity and validity of the assay. Finally, with GalTKO endothelia thrombus volume decreased by 60%, due primarily to a 58% reduction in adhesion (P < 0.0001 each); importantly, aggregation was only marginally affected (11% reduction, P < 0.0001). Conclusions/Significance This novel, high-throughput assay enabled dynamic modeling of whole-blood thrombosis on intact endothelium under physiologic conditions, and allowed mechanistic characterization of endothelial and platelet interactions. Applied to xenogeneic thrombosis, it enables future studies regarding the effect of modifying the porcine genotype on sheer-stress-dependent events that characterize xenograft injury. This in-vitro platform is likely to prove broadly useful to study thrombosis and endothelial interactions under dynamic physiologic conditions.
Collapse
Affiliation(s)
- Donald G Harris
- Division of General Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Prabhjot K Benipal
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Xiangfei Cheng
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Lars Burdorf
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Agnes M Azimzadeh
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Richard N Pierson
- Division of Cardiac Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America; Surgical Care Clinical Center, VA Maryland Health Care System, Baltimore, Maryland, United States of America
| |
Collapse
|
46
|
Arjunon S, Ardana PH, Saikrishnan N, Madhani S, Foster B, Glezer A, Yoganathan AP. Design of a pulsatile flow facility to evaluate thrombogenic potential of implantable cardiac devices. J Biomech Eng 2015; 137:045001. [PMID: 25587891 DOI: 10.1115/1.4029579] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Indexed: 11/08/2022]
Abstract
Due to expensive nature of clinical trials, implantable cardiac devices should first be extensively characterized in vitro. Prosthetic heart valves (PHVs), an important class of these devices, have been shown to be associated with thromboembolic complications. Although various in vitro systems have been designed to quantify blood-cell damage and platelet activation caused by nonphysiological hemodynamic shear stresses in these PHVs, very few systems attempt to characterize both blood damage and fluid dynamics aspects of PHVs in the same test system. Various numerical modeling methodologies are also evolving to simulate the structural mechanics, fluid mechanics, and blood damage aspects of these devices. This article presents a completely hemocompatible small-volume test-platform that can be used for thrombogenicity studies and experimental fluid mechanics characterization. Using a programmable piston pump to drive freshly drawn human blood inside a cylindrical column, the presented system can simulate various physiological and pathophysiological conditions in testing PHVs. The system includes a modular device-mounting chamber, and in this presented case, a 23 mm St. Jude Medical (SJM) Regents® mechanical heart valve (MHV) in aortic position was used as the test device. The system was validated for its capability to quantify blood damage by measuring blood damage induced by the tester itself (using freshly drawn whole human blood). Blood damage levels were ascertained through clinically relevant assays on human blood while fluid dynamics were characterized using time-resolved particle image velocimetry (PIV) using a blood-mimicking fluid. Blood damage induced by the tester itself, assessed through Thrombin-anti-Thrombin (TAT), Prothrombin factor 1.2 (PF1.2), and hemolysis (Drabkins assay), was within clinically accepted levels. The hydrodynamic performance of the tester showed consistent, repeatable physiological pressure and flow conditions. In addition, the system contains proximity sensors to accurately capture leaflet motion during the entire cardiac cycle. The PIV results showed skewing of the leakage jet, caused by the asymmetric closing of the two leaflets. All these results are critical to characterizing the blood damage and fluid dynamics characteristics of the SJM Regents® MHV, proving the utility of this tester as a precise system for assessing the hemodynamics and thrombogenicity for various PHVs.
Collapse
|
47
|
Ruggeri ZM, Mendolicchio GL. Interaction of von Willebrand factor with platelets and the vessel wall. Hamostaseologie 2015; 35:211-24. [PMID: 25612915 DOI: 10.5482/hamo-14-12-0081] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 12/09/2014] [Indexed: 01/19/2023] Open
Abstract
The initiation of thrombus formation at sites of vascular injury to secure haemostasis after tissue trauma requires the interaction of surface-exposed von Willebrand factor (VWF) with its primary platelet receptor, the glycoprotein (GP) Ib-IX-V complex. As an insoluble component of the extracellular matrix (ECM) of endothelial cells, VWF can directly initiate platelet adhesion. Circulating plasma VWF en-hances matrix VWF activity by binding to structures that become exposed to flowing blood, notably collagen type I and III in deeper layers of the vessel along with microfibrillar collagen type VI in the subendothelium. Moreover, plasma VWF is required to support platelet-to-platelet adhesion - i. e. aggregation - which promotes thrombus growth and consolidation. For these reasons, understanding how plasma VWF interaction with platelet receptors is regulated, particularly any distinctive features of GPIb binding to soluble as opposed to immobilized VWF, is of paramount importance in vascular biology. This brief review will highlight knowledge acquired and key problems that remain to be solved to elucidate fully the role of VWF in normal haemostasis and pathological thrombosis.
Collapse
Affiliation(s)
- Z M Ruggeri
- Zaverio M. Ruggeri, MD, The Scripps Research Institute, Maildrop: MEM 175, 10550 North Torrey Pines Road, La Jolla, California 92037, USA, Tel. 858/784 89 50, Fax 858/784 20 26, E-mail:
| | | |
Collapse
|
48
|
Goto S, Hasebe T, Takagi S. Platelets: Small in Size But Essential in the Regulation of Vascular Homeostasis – Translation From Basic Science to Clinical Medicine –. Circ J 2015; 79:1871-81. [DOI: 10.1253/circj.cj-14-1434] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Shinya Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine
- Department of Metabolic Disease Research Center, Tokai University Graduate School of Medicine
| | - Terumitsu Hasebe
- Department of Radiology, Tokai University Hachioji Hospital, Tokai University School of Medicine
| | - Shu Takagi
- Graduate School of Engineering, The University of Tokyo
| |
Collapse
|
49
|
Gifford SC, Spillane AM, Vignes SM, Shevkoplyas SS. Controlled incremental filtration: a simplified approach to design and fabrication of high-throughput microfluidic devices for selective enrichment of particles. LAB ON A CHIP 2014; 14:4496-505. [PMID: 25254358 PMCID: PMC4247995 DOI: 10.1039/c4lc00785a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The number of microfluidic strategies aimed at separating particles or cells of a specific size within a continuous flow system continues to grow. The wide array of biomedical and other applications that would benefit from successful development of such technology has motivated the extensive research in this area over the past 15 years. However, despite promising advancements in microfabrication capabilities, a versatile approach that is suitable for a large range of particle sizes and high levels of enrichment, with a volumetric throughput sufficient for large-scale applications, has yet to emerge. Here we describe a straightforward method that enables the rapid design of microfluidic devices that are capable of enriching/removing particles within a complex aqueous mixture, with an unprecedented range of potential cutoff diameter (below 1 μm to above 100 μm) and an easily scalable degree of enrichment/filtration (up to 10-fold and well beyond). A simplified model of a new approach to crossflow filtration - controlled incremental filtration - was developed and validated for its ability to generate microfluidic devices that efficiently separate particles on the order of 1-10 μm, with throughputs of tens of μL min(-1), without the use of a pump. Precise control of the amount of fluid incrementally diverted at each filtration "gap" of the device allows for the gap size (~20 μm) to be much larger than the particles of interest, while the simplicity of the model allows for many thousands of these filtration points to be readily incorporated into a desired device design. This new approach should enable truly high-throughput microfluidic particle-separation devices to be generated, even by users only minimally experienced in fluid mechanics and microfabrication techniques.
Collapse
Affiliation(s)
- Sean C Gifford
- Department of Biomedical Engineering, Tulane University, New Orleans, LA 70118, USA.
| | | | | | | |
Collapse
|
50
|
Tomita A, Tamura N, Nanazawa Y, Shiozaki S, Goto S. Development of virtual platelets implementing the functions of three platelet membrane proteins with different adhesive characteristics. J Atheroscler Thromb 2014; 22:201-10. [PMID: 25284441 DOI: 10.5551/jat.26203] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Computer simulation is a new method for understanding biological phenomena. In this report, we developed a simple platelet simulator representing platelet adhesion under blood flow conditions. METHODS We generated virtual platelets based on the functions of three key adhesive proteins: glycoprotein (GP) Ibα, GPIIb/IIIa and collagen receptors. The adhesive force between GPIbα and von Willebrand factor (VWF) was set to increase in association with increments in the fluid shear stress. GPIIb/IIIa acquires an adhesive force to bind with ligands only when platelets are activated following multiple GPIbα stimulation by VWF or collagen receptors. RESULTS Upon perfusion over the area of virtual endothelial injury, the virtual platelets adhered and became activated to form platelet thrombi. A total of 286/mm(2) of activated platelets was found to have accumulated downstream of the flow obstacle within 30 seconds, with 59/mm(2) platelets adhering upstream. The results obtained with the virtual model were consistent with those for real platelets in human blood in the presence of similarly shaped flow obstacles. CONCLUSIONS Our computer platelet simulator, which employs the functions of three key platelet membrane proteins, shows similar findings for adhesion in the presence and absence of blood flow obstacles.
Collapse
Affiliation(s)
- Aiko Tomita
- Department of Medicine (Cardiology), Tokai University School of Medicine
| | | | | | | | | |
Collapse
|