1
|
Cacho-Navas C, López-Pujante C, Reglero-Real N, Colás-Algora N, Cuervo A, Conesa JJ, Barroso S, de Rivas G, Ciordia S, Paradela A, D'Agostino G, Manzo C, Feito J, Andrés G, Molina-Jiménez F, Majano P, Correas I, Carazo JM, Nourshargh S, Huch M, Millán J. ICAM-1 nanoclusters regulate hepatic epithelial cell polarity by leukocyte adhesion-independent control of apical actomyosin. eLife 2024; 12:RP89261. [PMID: 38597186 PMCID: PMC11006420 DOI: 10.7554/elife.89261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024] Open
Abstract
Epithelial intercellular adhesion molecule (ICAM)-1 is apically polarized, interacts with, and guides leukocytes across epithelial barriers. Polarized hepatic epithelia organize their apical membrane domain into bile canaliculi and ducts, which are not accessible to circulating immune cells but that nevertheless confine most of ICAM-1. Here, by analyzing ICAM-1_KO human hepatic cells, liver organoids from ICAM-1_KO mice and rescue-of-function experiments, we show that ICAM-1 regulates epithelial apicobasal polarity in a leukocyte adhesion-independent manner. ICAM-1 signals to an actomyosin network at the base of canalicular microvilli, thereby controlling the dynamics and size of bile canalicular-like structures. We identified the scaffolding protein EBP50/NHERF1/SLC9A3R1, which connects membrane proteins with the underlying actin cytoskeleton, in the proximity interactome of ICAM-1. EBP50 and ICAM-1 form nano-scale domains that overlap in microvilli, from which ICAM-1 regulates EBP50 nano-organization. Indeed, EBP50 expression is required for ICAM-1-mediated control of BC morphogenesis and actomyosin. Our findings indicate that ICAM-1 regulates the dynamics of epithelial apical membrane domains beyond its role as a heterotypic cell-cell adhesion molecule and reveal potential therapeutic strategies for preserving epithelial architecture during inflammatory stress.
Collapse
Affiliation(s)
| | | | - Natalia Reglero-Real
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of LondonLondonUnited Kingdom
| | | | - Ana Cuervo
- Centro Nacional de Biotecnologia (CSIC)MadridSpain
| | | | - Susana Barroso
- Centro de Biologia Molecular Severo Ochoa, CSIC-UAMMadridSpain
| | - Gema de Rivas
- Centro de Biologia Molecular Severo Ochoa, CSIC-UAMMadridSpain
| | | | | | | | - Carlo Manzo
- Facultat de Ciències, Tecnologia i Enginyeries, Universitat de Vic – Universitat Central de Catalunya (UVic-UCC)VicSpain
| | - Jorge Feito
- Servicio de Anatomía Patológica, Hospital Universitario de SalamancaSalamancaSpain
| | - Germán Andrés
- Centro de Biologia Molecular Severo Ochoa, CSIC-UAMMadridSpain
| | - Francisca Molina-Jiménez
- Molecular Biology Unit, Hospital Universitario de la PrincesaMadridSpain
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa)MadridSpain
| | - Pedro Majano
- Molecular Biology Unit, Hospital Universitario de la PrincesaMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)MadridSpain
- Department of Cellular Biology, Universidad Complutense de MadridMadridSpain
| | - Isabel Correas
- Centro de Biologia Molecular Severo Ochoa, CSIC-UAMMadridSpain
| | | | - Sussan Nourshargh
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of LondonLondonUnited Kingdom
| | - Meritxell Huch
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Jaime Millán
- Centro de Biologia Molecular Severo Ochoa, CSIC-UAMMadridSpain
| |
Collapse
|
2
|
Kenny M, Pollitt AY, Patil S, Hiebner DW, Smolenski A, Lakic N, Fisher R, Alsufyani R, Lickert S, Vogel V, Schoen I. Contractility defects hinder glycoprotein VI-mediated platelet activation and affect platelet functions beyond clot contraction. Res Pract Thromb Haemost 2024; 8:102322. [PMID: 38379711 PMCID: PMC10877441 DOI: 10.1016/j.rpth.2024.102322] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 12/23/2023] [Accepted: 01/03/2024] [Indexed: 02/22/2024] Open
Abstract
Background Active and passive biomechanical properties of platelets contribute substantially to thrombus formation. Actomyosin contractility drives clot contraction required for stabilizing the hemostatic plug. Impaired contractility results in bleeding but is difficult to detect using platelet function tests. Objectives To determine how diminished myosin activity affects platelet functions, including and beyond clot contraction. Methods Using the myosin IIA-specific pharmacologic inhibitor blebbistatin, we modulated myosin activity in platelets from healthy donors and systematically characterized platelet responses at various levels of inhibition by interrogating distinct platelet functions at each stage of thrombus formation using a range of complementary assays. Results Partial myosin IIA inhibition neither affected platelet von Willebrand factor interactions under arterial shear nor platelet spreading and cytoskeletal rearrangements on fibrinogen. However, it impacted stress fiber formation and the nanoarchitecture of cell-matrix adhesions, drastically reducing and limiting traction forces. Higher blebbistatin concentrations impaired platelet adhesion under flow, altered mechanosensing at lamellipodia edges, and eliminated traction forces without affecting platelet spreading, α-granule secretion, or procoagulant platelet formation. Unexpectedly, myosin IIA inhibition reduced calcium influx, dense granule secretion, and platelet aggregation downstream of glycoprotein (GP)VI and limited the redistribution of GPVI on the cell membrane, whereas aggregation induced by adenosine diphosphate or arachidonic acid was unaffected. Conclusion Our findings highlight the importance of both active contractile and passive crosslinking roles of myosin IIA in the platelet cytoskeleton. They support the hypothesis that highly contractile platelets are needed for hemostasis and further suggest a supportive role for myosin IIA in GPVI signaling.
Collapse
Affiliation(s)
- Martin Kenny
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Alice Y. Pollitt
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Smita Patil
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Dishon W. Hiebner
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Albert Smolenski
- School of Medicine, Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| | - Natalija Lakic
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Robert Fisher
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Reema Alsufyani
- School of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Sebastian Lickert
- Department of Health Sciences and Technologies, ETH Zurich, Zurich, Switzerland
| | - Viola Vogel
- Department of Health Sciences and Technologies, ETH Zurich, Zurich, Switzerland
| | - Ingmar Schoen
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
3
|
Zelená A, Blumberg J, Probst D, Gerasimaitė R, Lukinavičius G, Schwarz US, Köster S. Force generation in human blood platelets by filamentous actomyosin structures. Biophys J 2023; 122:3340-3353. [PMID: 37475214 PMCID: PMC10465724 DOI: 10.1016/j.bpj.2023.07.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 02/11/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023] Open
Abstract
Blood platelets are central elements of the blood clotting response after wounding. Upon vessel damage, they bind to the surrounding matrix and contract the forming thrombus, thus helping to restore normal blood circulation. The hemostatic function of platelets is directly connected to their mechanics and cytoskeletal organization. The reorganization of the platelet cytoskeleton during spreading occurs within minutes and leads to the formation of contractile actomyosin bundles, but it is not known if there is a direct correlation between the emerging actin structures and the force field that is exerted to the environment. In this study, we combine fluorescence imaging of the actin structures with simultaneous traction force measurements in a time-resolved manner. In addition, we image the final states with superresolution microscopy. We find that both the force fields and the cell shapes have clear geometrical patterns defined by stress fibers. Force generation is localized in a few hotspots, which appear early during spreading, and, in the mature state, anchor stress fibers in focal adhesions. Moreover, we show that, for a gel stiffness in the physiological range, force generation is a very robust mechanism and we observe no systematic dependence on the amount of added thrombin in solution or fibrinogen coverage on the substrate, suggesting that force generation after platelet activation is a threshold phenomenon that ensures reliable thrombus contraction in diverse environments.
Collapse
Affiliation(s)
- Anna Zelená
- Institute for X-Ray Physics, University of Göttingen, Göttingen, Germany
| | - Johannes Blumberg
- Institute for Theoretical Physics, University of Heidelberg, Heidelberg, Germany; BioQuant-Center for Quantitative Biology, University of Heidelberg, Heidelberg, Germany
| | - Dimitri Probst
- Institute for Theoretical Physics, University of Heidelberg, Heidelberg, Germany; BioQuant-Center for Quantitative Biology, University of Heidelberg, Heidelberg, Germany
| | - Rūta Gerasimaitė
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | | | - Ulrich S Schwarz
- Institute for Theoretical Physics, University of Heidelberg, Heidelberg, Germany; BioQuant-Center for Quantitative Biology, University of Heidelberg, Heidelberg, Germany.
| | - Sarah Köster
- Institute for X-Ray Physics, University of Göttingen, Göttingen, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| |
Collapse
|
4
|
Severin S, Consonni A, Chicanne G, Allart S, Payrastre B, Gratacap MP. SHIP1 Controls Internal Platelet Contraction and α IIbβ 3 Integrin Dynamics in Early Platelet Activation. Int J Mol Sci 2023; 24:ijms24020958. [PMID: 36674478 PMCID: PMC9860818 DOI: 10.3390/ijms24020958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 01/06/2023] Open
Abstract
The Src homology 2 domain-containing inositol 5-phosphatase 1 (SHIP1) is known to dephosphorylate PtdIns(3,4,5)P3 into PtdIns(3,4)P2 and to interact with several signaling proteins though its docking functions. It has been shown to negatively regulate platelet adhesion and spreading on a fibrinogen surface and to positively regulate thrombus growth. In the present study, we have investigated its role during the early phase of platelet activation. Using confocal-based morphometric analysis, we found that SHIP1 is involved in the regulation of cytoskeletal organization and internal contractile activity in thrombin-activated platelets. The absence of SHIP1 has no significant impact on thrombin-induced Akt or Erk1/2 activation, but it selectively affects the RhoA/Rho-kinase pathway and myosin IIA relocalization to the cytoskeleton. SHIP1 interacts with the spectrin-based membrane skeleton, and its absence induces a loss of sustained association of integrins to this network together with a decrease in αIIbβ3 integrin clustering following thrombin stimulation. This αIIbβ3 integrin dynamics requires the contractile cytoskeleton under the control of SHIP1. RhoA activation, internal platelet contraction, and membrane skeleton integrin association were insensitive to the inhibition of PtdIns(3,4,5)P3 synthesis or SHIP1 phosphatase activity, indicating a role for the docking properties of SHIP1 in these processes. Altogether, our data reveal a lipid-independent function for SHIP1 in the regulation of the contractile cytoskeleton and integrin dynamics in platelets.
Collapse
Affiliation(s)
- Sonia Severin
- Institut des Maladies Métabolique et Cardiovasculaire (I2MC), Inserm and Université Toulouse III Paul-Sabatier (UMR-1297), 1 Avenue J. Poulhes, CEDEX 4, 31432 Toulouse, France
- Correspondence: (S.S.); (M.-P.G.); Tel.: +33-5-31-22-41-43 (S.S.); +33-5-31-22-41-50 (M.-P.G.)
| | - Alessandra Consonni
- Institut des Maladies Métabolique et Cardiovasculaire (I2MC), Inserm and Université Toulouse III Paul-Sabatier (UMR-1297), 1 Avenue J. Poulhes, CEDEX 4, 31432 Toulouse, France
- Laboratory of Biochemistry, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy
| | - Gaëtan Chicanne
- Institut des Maladies Métabolique et Cardiovasculaire (I2MC), Inserm and Université Toulouse III Paul-Sabatier (UMR-1297), 1 Avenue J. Poulhes, CEDEX 4, 31432 Toulouse, France
| | - Sophie Allart
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), Université Toulouse III Paul-Sabatier and Inserm (UMR-1291) and CNRS (UMR-5051), Centre Hospitalier Universitaire Purpan, CEDEX 3, 31024 Toulouse, France
| | - Bernard Payrastre
- Institut des Maladies Métabolique et Cardiovasculaire (I2MC), Inserm and Université Toulouse III Paul-Sabatier (UMR-1297), 1 Avenue J. Poulhes, CEDEX 4, 31432 Toulouse, France
- Laboratoire d’Hématologie, Centre de Référence des Pathologies Plaquettaires, Centre Hospitalier Universitaire Rangueil, CEDEX 4, 31432 Toulouse, France
| | - Marie-Pierre Gratacap
- Institut des Maladies Métabolique et Cardiovasculaire (I2MC), Inserm and Université Toulouse III Paul-Sabatier (UMR-1297), 1 Avenue J. Poulhes, CEDEX 4, 31432 Toulouse, France
- Correspondence: (S.S.); (M.-P.G.); Tel.: +33-5-31-22-41-43 (S.S.); +33-5-31-22-41-50 (M.-P.G.)
| |
Collapse
|
5
|
Li Y, Xin G, Li S, Dong Y, Zhu Y, Yu X, Wan C, Li F, Wei Z, Wang Y, Zhang K, Chen Q, Niu H, Huang W. PD-L1 Regulates Platelet Activation and Thrombosis via Caspase-3/GSDME Pathway. Front Pharmacol 2022; 13:921414. [PMID: 35784685 PMCID: PMC9240427 DOI: 10.3389/fphar.2022.921414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/25/2022] [Indexed: 11/24/2022] Open
Abstract
Platelets play a central role in hemostasis and thrombosis, regulating the occurrence and development of thrombotic diseases, including ischemic stroke. Programmed death ligand 1 (PD-L1) has recently been detected in platelet, while the function of PD-L1 in platelets remain elusive. Our data reveal a novel mechanism for the role of PD-L1 on platelet activation and arterial thrombosis. PD-L1 knockout does not affect platelet morphology, count, and mean volume under homeostasis and without risk of bleeding, which inhibits platelet activation by suppressing outside-in-activation of integrin by downregulating the Caspase-3/GSDME pathway. Platelet adoptive transfer experiments demonstrate that PD-L1 knockout inhibits thrombosis. And the absence of PD-L1 improves ischemic stroke severity and increases mice survival. Immunohistochemical staining of the internal structure of the thrombus proves that PD-L1 enhances the seriousness of the thrombus by inhibiting platelet activation. This work reveals a regulatory role of PD-L1 on platelet activation and thrombosis while providing novel platelet intervention strategies to prevent thrombosis.
Collapse
|
6
|
Teeraratkul C, Mukherjee D. Microstructure aware modeling of biochemical transport in arterial blood clots. J Biomech 2021; 127:110692. [PMID: 34479090 DOI: 10.1016/j.jbiomech.2021.110692] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 07/14/2021] [Accepted: 08/09/2021] [Indexed: 01/29/2023]
Abstract
Flow-mediated transport of biochemical species is central to thrombotic phenomena. Comprehensive three-dimensional modeling of flow-mediated transport around realistic macroscale thrombi poses challenges owing to their arbitrary heterogeneous microstructure. Here, we develop a microstructure aware model for species transport within and around a macroscale thrombus by devising a custom preconditioned fictitious domain formulation for thrombus-hemodynamics interactions, and coupling it with a fictitious domain advection-diffusion formulation for transport. Microstructural heterogeneities are accounted through a hybrid discrete particle-continuum approach for the thrombus interior. We present systematic numerical investigations on unsteady arterial flow within and around a three-dimensional macroscale thrombus; demonstrate the formation of coherent flow structures around the thrombus which organize advective transport; illustrate the role of the permeation processes at the thrombus boundary and subsequent intra-thrombus transport; and characterize species transport from bulk flow to the thrombus boundary and vice versa.
Collapse
Affiliation(s)
- Chayut Teeraratkul
- Paul M Rady Department of Mechanical Engineering, University of Colorado Boulder, United States of America.
| | - Debanjan Mukherjee
- Paul M Rady Department of Mechanical Engineering, University of Colorado Boulder, United States of America.
| |
Collapse
|
7
|
Ngo ATP, Parra-Izquierdo I, Aslan JE, McCarty OJT. Rho GTPase regulation of reactive oxygen species generation and signalling in platelet function and disease. Small GTPases 2021; 12:440-457. [PMID: 33459160 DOI: 10.1080/21541248.2021.1878001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Platelets are master regulators and effectors of haemostasis with increasingly recognized functions as mediators of inflammation and immune responses. The Rho family of GTPase members Rac1, Cdc42 and RhoA are known to be major components of the intracellular signalling network critical to platelet shape change and morphological dynamics, thus playing a major role in platelet spreading, secretion and thrombus formation. Initially linked to the regulation of actomyosin contraction and lamellipodia formation, recent reports have uncovered non-canonical functions of platelet RhoGTPases in the regulation of reactive oxygen species (ROS), where intrinsically generated ROS modulate platelet function and contribute to thrombus formation. Platelet RhoGTPases orchestrate oxidative processes and cytoskeletal rearrangement in an interconnected manner to regulate intracellular signalling networks underlying platelet activity and thrombus formation. Herein we review our current knowledge of the regulation of platelet ROS generation by RhoGTPases and their relationship with platelet cytoskeletal reorganization, activation and function.
Collapse
Affiliation(s)
- Anh T P Ngo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Ivan Parra-Izquierdo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA.,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph E Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA.,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA.,Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
8
|
Teeraratkul C, Irwin Z, Shadden SC, Mukherjee D. Computational investigation of blood flow and flow-mediated transport in arterial thrombus neighborhood. Biomech Model Mechanobiol 2021; 20:701-715. [PMID: 33438148 DOI: 10.1007/s10237-020-01411-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 12/09/2020] [Indexed: 12/19/2022]
Abstract
A pathologically formed blood clot or thrombus is central to major cardiovascular diseases like heart attack and stroke. Detailed quantitative evaluation of flow and flow-mediated transport processes in the thrombus neighborhood within large artery hemodynamics is crucial for understanding disease progression and assessing treatment efficacy. This, however, remains a challenging task owing to the complexity of pulsatile viscous flow interactions with arbitrary shape and heterogeneous microstructure of realistic thrombi. Here, we address this challenge by conducting a systematic parametric simulation-based study on characterizing unsteady hemodynamics and flow-mediated transport in the neighborhood of an arterial thrombus. We use a hybrid particle-continuum-based finite element approach to handle arbitrary thrombus shape and microstructural variations. Results from a cohort of 50 different unsteady flow scenarios are presented, including unsteady vortical structures, pressure gradient across the thrombus boundary, finite time Lyapunov exponents, and dynamic coherent structures that organize advective transport. We clearly illustrate the combined influence of three key parameters-thrombus shape, microstructure, and extent of wall disease-in terms of: (a) determining hemodynamic features in the thrombus neighborhood and (b) governing the balance between advection, permeation, and diffusion to regulate transport processes in the thrombus neighborhood.
Collapse
Affiliation(s)
- Chayut Teeraratkul
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, United States of America
| | - Zachariah Irwin
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, United States of America
| | - Shawn C Shadden
- Department of Mechanical Engineering, University of California, Berkeley, United States of America
| | - Debanjan Mukherjee
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, United States of America.
| |
Collapse
|
9
|
Huang K, Li Z, Su T, Shen D, Hu S, Cheng K. Bispecific Antibody Therapy for Effective Cardiac Repair through Redirection of Endogenous Stem Cells. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Ke Huang
- Department of Molecular Biomedical Sciences North Carolina University Raleigh NC 27607 USA
| | - Zhenhua Li
- Department of Molecular Biomedical Sciences North Carolina University Raleigh NC 27607 USA
- Joint Department of Biomedical Engineering University of North Carolina at Chapel Hill and North Carolina State University Raleigh NC 27695 USA
| | - Teng Su
- Department of Molecular Biomedical Sciences North Carolina University Raleigh NC 27607 USA
- Joint Department of Biomedical Engineering University of North Carolina at Chapel Hill and North Carolina State University Raleigh NC 27695 USA
| | - Deliang Shen
- Department of Cardiology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450052 China
| | - Shiqi Hu
- Department of Molecular Biomedical Sciences North Carolina University Raleigh NC 27607 USA
- Joint Department of Biomedical Engineering University of North Carolina at Chapel Hill and North Carolina State University Raleigh NC 27695 USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences North Carolina University Raleigh NC 27607 USA
- Joint Department of Biomedical Engineering University of North Carolina at Chapel Hill and North Carolina State University Raleigh NC 27695 USA
- Division of Pharmacoengineering and Molecular Pharmaceutics Eshelman School of Pharmacy University of North Carolina at Chapel Hill Chapel Hill NC 27599 USA
| |
Collapse
|
10
|
Contractile forces in platelet aggregates under microfluidic shear gradients reflect platelet inhibition and bleeding risk. Nat Commun 2019; 10:1204. [PMID: 30867419 PMCID: PMC6416331 DOI: 10.1038/s41467-019-09150-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 02/13/2019] [Indexed: 12/21/2022] Open
Abstract
Platelets contract forcefully after their activation, contributing to the strength and stability of platelet aggregates and fibrin clots during blood coagulation. Viscoelastic approaches can be used to assess platelet-induced clot strengthening, but they require thrombin and fibrin generation and are unable to measure platelet forces directly. Here, we report a rapid, microfluidic approach for measuring the contractile force of platelet aggregates for the detection of platelet dysfunction. We find that platelet forces are significantly reduced when blood samples are treated with inhibitors of myosin, GPIb-IX-V, integrin αIIbβ3, P2Y12, or thromboxane generation. Clinically, we find that platelet forces are measurably lower in cardiology patients taking aspirin. We also find that measuring platelet forces can identify Emergency Department trauma patients who subsequently require blood transfusions. Together, these findings indicate that microfluidic quantification of platelet forces may be a rapid and useful approach for monitoring both antiplatelet therapy and traumatic bleeding risk. Platelet aggregates generate contractile forces that contribute to their cohesion and adhesion. Here, Ting et al. develop a microfluidic device to measure contractile forces generated by platelet aggregates, and find it can detect the response of platelets to pharmacological agents and predict bleeding risk in trauma patients.
Collapse
|
11
|
|
12
|
Inhibition of myosin IIA–actin interaction prevents ischemia/reperfusion induced cardiomyocytes apoptosis through modulating PINK1/Parkin pathway and mitochondrial fission. Int J Cardiol 2018; 271:211-218. [DOI: 10.1016/j.ijcard.2018.04.079] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 04/09/2018] [Accepted: 04/18/2018] [Indexed: 11/21/2022]
|
13
|
Paknikar AK, Eltzner B, Köster S. Direct characterization of cytoskeletal reorganization during blood platelet spreading. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 144:166-176. [PMID: 29843920 DOI: 10.1016/j.pbiomolbio.2018.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/15/2018] [Accepted: 05/04/2018] [Indexed: 11/18/2022]
Abstract
Blood platelets are the key cellular players in blood clotting and thus of great biomedical importance. While spreading at the site of injury, they reorganize their cytoskeleton within minutes and assume a flat appearance. As platelets possess no nucleus, many standard methods for visualizing cytoskeletal components by means of fluorescence tags fail. Here we employ silicon-rhodamine actin and tubulin probes for imaging these important proteins in a time-resolved manner. We find two distinct timescales for platelet spread area development and for cytoskeletal reorganization, indicating that although cell spreading is most likely associated with actin polymerization at the cell edges, distinct, stress-fiber-like actin structures within the cell, which may be involved in the generation of contractile forces, form on their own timescale. Following microtubule dynamics allows us to distinguish the role of myosin, microtubules and actin during early spreading.
Collapse
Affiliation(s)
- Aishwarya K Paknikar
- Institute for X-Ray Physics, University of Goettingen, Göttingen, 37077, Germany
| | - Benjamin Eltzner
- Institute for Mathematical Stochastics, University of Goettingen, Göttingen, 37077, Germany
| | - Sarah Köster
- Institute for X-Ray Physics, University of Goettingen, Göttingen, 37077, Germany; DZHK (German Center for Cardiovascular Research), partner site Göttingen, Germany.
| |
Collapse
|
14
|
Atkinson L, Yusuf MZ, Aburima A, Ahmed Y, Thomas SG, Naseem KM, Calaminus SDJ. Reversal of stress fibre formation by Nitric Oxide mediated RhoA inhibition leads to reduction in the height of preformed thrombi. Sci Rep 2018; 8:3032. [PMID: 29445102 PMCID: PMC5813033 DOI: 10.1038/s41598-018-21167-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/24/2018] [Indexed: 01/10/2023] Open
Abstract
Evidence has emerged to suggest that thrombi are dynamic structures with distinct areas of differing platelet activation and inhibition. We hypothesised that Nitric oxide (NO), a platelet inhibitor, can modulate the actin cytoskeleton reversing platelet spreading, and therefore reduce the capability of thrombi to withstand a high shear environment. Our data demonstrates that GSNO, DEANONOate, and a PKG-activating cGMP analogue reversed stress fibre formation and increased actin nodule formation in adherent platelets. This effect is sGC dependent and independent of ADP and thromboxanes. Stress fibre formation is a RhoA dependent process and NO induced RhoA inhibition, however, it did not phosphorylate RhoA at ser188 in spread platelets. Interestingly NO and PGI2 synergise to reverse stress fibre formation at physiologically relevant concentrations. Analysis of high shear conditions indicated that platelets activated on fibrinogen, induced stress fibre formation, which was reversed by GSNO treatment. Furthermore, preformed thrombi on collagen post perfused with GSNO had a 30% reduction in thrombus height in comparison to the control. This study demonstrates that NO can reverse key platelet functions after their initial activation and identifies a novel mechanism for controlling excessive thrombosis.
Collapse
Affiliation(s)
- L Atkinson
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK
| | - M Z Yusuf
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK
| | - A Aburima
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK
| | - Y Ahmed
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK
| | - S G Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - K M Naseem
- Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, UK
| | - S D J Calaminus
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK.
| |
Collapse
|
15
|
Platelet integrins exhibit anisotropic mechanosensing and harness piconewton forces to mediate platelet aggregation. Proc Natl Acad Sci U S A 2017; 115:325-330. [PMID: 29269394 DOI: 10.1073/pnas.1710828115] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Platelet aggregation at the site of vascular injury is essential in clotting. During this process, platelets are bridged by soluble fibrinogen that binds surface integrin receptors. One mystery in the mechanism of platelet aggregation pertains to how resting platelets ignore soluble fibrinogen, the third most abundant protein in the bloodstream, and yet avidly bind immobile fibrinogen on the surface of other platelets at the primary injury site. We speculate that platelet integrins are mechanosensors that test their ligands across the platelet-platelet synapse. To investigate this model, we interrogate human platelets using approaches that include the supported lipid bilayer platform as well as DNA tension sensor technologies. Experiments suggest that platelet integrins require lateral forces to mediate platelet-platelet interactions. Mechanically labile ligands dampen platelet activation, and the onset of piconewton integrin tension coincides with calcium flux. Activated platelets display immobilized fibrinogen on their surface, thus mediating further recruitment of resting platelets. The distribution of integrin tension was shown to be spatially regulated through two myosin-signaling pathways, myosin light chain kinase and Rho-associated kinase. Finally, we discovered that the termination of integrin tension is coupled with the exposure of phosphatidylserine. Our work reveals the highest spatial and temporal resolution maps of platelet integrin mechanics and its role in platelet aggregation, suggesting that platelets are physical substrates for one another that establish mechanical feedback loops of activation. The results are reminiscent of mechanical regulation of the T-cell receptor, E-cadherin, and Notch pathways, suggesting a common feature for signaling at cell junctions.
Collapse
|
16
|
Endogenous fibrinolysis facilitates clot retraction in vivo. Blood 2017; 130:2453-2462. [DOI: 10.1182/blood-2017-06-789032] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/18/2017] [Indexed: 12/22/2022] Open
Abstract
Key Points
Localized vascular injury with thrombin microinjection produces a fibrin network that undergoes myosin IIa–dependent retraction in vivo. Using this model, we demonstrate that endogenous fibrinolysis promotes fibrin clot retraction.
Collapse
|
17
|
Egot M, Kauskot A, Lasne D, Gaussem P, Bachelot-Loza C. Biphasic myosin II light chain activation during clot retraction. Thromb Haemost 2017; 110:1215-22. [DOI: 10.1160/th13-04-0335] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 08/05/2013] [Indexed: 12/13/2022]
Abstract
SummaryClot retraction is an essential step during primary haemostasis, thereby promoting thrombus stability and wound healing. Integrin αIIbβ3 plays a critical role in clot retraction, by inducing acto-myosin interactions that allow platelet cytoskeleton reorganisation. However, the signalling pathways that lead to clot retraction are still misunderstood. In this study, we report the first data on the kinetics of myosin II light chain (MLC) phosphorylation during clot retraction. We found an early phosphorylation peak followed by a second peak. By using specific inhibitors of kinases and small G proteins, we showed that MLC kinase (MLCK), RhoA/ROCK, and Rac-1 were involved in clot retraction and in the early MLC phosphorylation peak. Only Rac-1 and actin polymerisation, controlled by outside-in signalling, were crucial to the second MLC phosphorylation peak.
Collapse
|
18
|
Modeling blood flow around a thrombus using a hybrid particle–continuum approach. Biomech Model Mechanobiol 2017; 17:645-663. [DOI: 10.1007/s10237-017-0983-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 11/09/2017] [Indexed: 10/18/2022]
|
19
|
Feghhi S, Tooley WW, Sniadecki NJ. Nonmuscle Myosin IIA Regulates Platelet Contractile Forces Through Rho Kinase and Myosin Light-Chain Kinase. J Biomech Eng 2017; 138:2546290. [PMID: 27548633 DOI: 10.1115/1.4034489] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Indexed: 11/08/2022]
Abstract
Platelet contractile forces play a major role in clot retraction and help to hold hemostatic clots against the vessel wall. Platelet forces are produced by its cytoskeleton, which is composed of actin and nonmuscle myosin filaments. In this work, we studied the role of Rho kinase, myosin light-chain kinase, and myosin in the generation of contractile forces by using pharmacological inhibitors and arrays of flexible microposts to measure platelet forces. When platelets were seeded onto microposts, they formed aggregates on the tips of the microposts. Forces produced by the platelets in the aggregates were measured by quantifying the deflection of the microposts, which bent in proportion to the force of the platelets. Platelets were treated with small molecule inhibitors of myosin activity: Y-27632 to inhibit the Rho kinase (ROCK), ML-7 to inhibit myosin light-chain kinase (MLCK), and blebbistatin to inhibit myosin ATPase activity. ROCK inhibition reduced platelet forces, demonstrating the importance of the assembly of actin and myosin phosphorylation in generating contractile forces. Similarly, MLCK inhibition caused weaker platelet forces, which verifies that myosin phosphorylation is needed for force generation in platelets. Platelets treated with blebbistatin also had weaker forces, which indicates that myosin's ATPase activity is necessary for platelet forces. Our studies demonstrate that myosin ATPase activity and the regulation of actin-myosin assembly by ROCK and MLCK are needed for the generation of platelet forces. Our findings illustrate and explain the importance of myosin for clot compaction in hemostasis and thrombosis.
Collapse
|
20
|
Force-activatable biosensor enables single platelet force mapping directly by fluorescence imaging. Biosens Bioelectron 2017; 100:192-200. [PMID: 28915383 DOI: 10.1016/j.bios.2017.09.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/25/2017] [Accepted: 09/05/2017] [Indexed: 11/23/2022]
Abstract
Integrin-transmitted cellular forces are critical for platelet adhesion, activation, aggregation and contraction during hemostasis and thrombosis. Measuring and mapping single platelet forces are desired in both research and clinical applications. Conventional force-to-strain based cell traction force microscopies have low resolution which is not ideal for cellular force mapping in small platelets. To enable platelet force mapping with submicron resolution, we developed a force-activatable biosensor named integrative tension sensor (ITS) which directly converts molecular tensions to fluorescent signals, therefore enabling cellular force mapping directly by fluorescence imaging. With ITS, we mapped cellular forces in single platelets at 0.4µm resolution. We found that platelet force distribution has strong polarization which is sensitive to treatment with the anti-platelet drug tirofiban, suggesting that the ITS force map can report anti-platelet drug efficacy. The ITS also calibrated integrin molecular tensions in platelets and revealed two distinct tension levels: 12-54 piconewton (nominal values) tensions generated during platelet adhesion and tensions above 54 piconewton generated during platelet contraction. Overall, the ITS is a powerful biosensor for the study of platelet mechanobiology, and holds great potential in antithrombotic drug development and assessing platelet activity in health and disease.
Collapse
|
21
|
Zhai K, Zheng J, Tang Y, Li F, Lv Y, Zhang Y, Gao Z, Qi J, Yu B, Kou J. The saponin D39 blocks dissociation of non-muscular myosin heavy chain IIA from TNF receptor 2, suppressing tissue factor expression and venous thrombosis. Br J Pharmacol 2017; 174:2818-2831. [PMID: 28547925 PMCID: PMC5554322 DOI: 10.1111/bph.13885] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 04/27/2017] [Accepted: 05/08/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Non-muscular myosin heavy chain IIA (NMMHC IIA) plays a key role in tissue factor expression and venous thrombosis. Natural products might inhibit thrombosis through effects on NMMHC IIA. Here, we have shown that a natural saponin, D39, from Liriope muscari exerted anti-thrombotic activity in vivo, by targeting NMMHC IIA. EXPERIMENTAL APPROACH Expression and activity of tissue factor in endothelial cells were analysed in vitro by Western blot and simplified chromogenic assays. Interactions between D39 and NMMHC IIA were assessed by serial affinity chromatography and molecular docking analysis. D39-dependent interactions between NMMHC IIA and TNF receptor 2 (TNFR2) were measured by immunofluorescence, co-immunoprecipitation and proximity ligation assays. Anti-thrombotic activity of D39 in vivo was evaluated with a model of inferior vena cava ligation injury in mice. KEY RESULTS D39 inhibited tissue factor expression and procoagulant activities in HUVECs and decreased thrombus weight in inferior vena cava-ligated mice dose-dependently. Serial affinity chromatography and molecular docking analysis suggested that D39 bound to NMMHC IIA. In HEK293T cells, D39 inhibited tissue factor expression evoked by NMMHC IIA overexpression. This effect was blocked by NMMHC IIA knockdown in HUVECs. D39 inhibited dissociation of NMMHC IIA from TNFR2, which subsequently modulated the Akt/GSK3β-NF-κB signalling pathways. CONCLUSIONS AND IMPLICATIONS D39 inhibited tissue factor expression and thrombus formation by modulating the Akt/GSK3β and NF-κB signalling pathways through NMMHC IIA. We identified a new natural product that targeted NMMHC IIA, as a potential treatment for thrombotic disorders and other vasculopathies.
Collapse
Affiliation(s)
- Ke‐feng Zhai
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
- Institute of Pharmaceutical Biotechnology, School of Biological and Food EngineeringSuzhou UniversitySuzhouChina
| | - Jin‐rong Zheng
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| | - You‐mei Tang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| | - Fang Li
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| | - Yan‐ni Lv
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| | - Yuan‐yuan Zhang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| | - Zhen Gao
- Department of Medicine‐Ather&LipoBaylor Colledge of MedicineHoustonTXUSA
| | - Jin Qi
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| | - Bo‐yang Yu
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| | - Jun‐ping Kou
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| |
Collapse
|
22
|
Prostacyclin reverses platelet stress fibre formation causing platelet aggregate instability. Sci Rep 2017; 7:5582. [PMID: 28717253 PMCID: PMC5514131 DOI: 10.1038/s41598-017-05817-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/02/2017] [Indexed: 01/10/2023] Open
Abstract
Prostacyclin (PGI2) modulates platelet activation to regulate haemostasis. Evidence has emerged to suggest that thrombi are dynamic structures with distinct areas of differing platelet activation. It was hypothesised that PGI2 could reverse platelet spreading by actin cytoskeletal modulation, leading to reduced capability of platelet aggregates to withstand a high shear environment. Our data demonstrates that post-flow of PGI2 over activated and spread platelets on fibrinogen, identified a significant reduction in platelet surface area under high shear. Exploration of the molecular mechanisms underpinning this effect revealed that PGI2 reversed stress fibre formation in adherent platelets, reduced platelet spreading, whilst simultaneously promoting actin nodule formation. The effects of PGI2 on stress fibres were mimicked by the adenylyl cyclase activator forskolin and prevented by inhibitors of protein kinase A (PKA). Stress fibre formation is a RhoA dependent process and we found that treatment of adherent platelets with PGI2 caused inhibitory phosphorylation of RhoA, reduced RhoA GTP-loading and reversal of myosin light chain phosphorylation. Phospho-RhoA was localised in actin nodules with PKA type II and a number of other phosphorylated PKA substrates. This study demonstrates that PGI2 can reverse key platelet functions after their initial activation and identifies a novel mechanism for controlling thrombosis.
Collapse
|
23
|
Seifert J, Rheinlaender J, Lang F, Gawaz M, Schäffer TE. Thrombin-induced cytoskeleton dynamics in spread human platelets observed with fast scanning ion conductance microscopy. Sci Rep 2017; 7:4810. [PMID: 28684746 PMCID: PMC5500533 DOI: 10.1038/s41598-017-04999-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 05/19/2017] [Indexed: 02/08/2023] Open
Abstract
Platelets are small anucleate blood cells involved in haemostasis. Platelet activation, caused by agonists such as thrombin or by contact with the extracellular matrix, leads to platelet adhesion, aggregation, and coagulation. Activated platelets undergo shape changes, adhere, and spread at the site of injury to form a blood clot. We investigated the morphology and morphological dynamics of human platelets after complete spreading using fast scanning ion conductance microscopy (SICM). In contrast to unstimulated platelets, thrombin-stimulated platelets showed increased morphological activity after spreading and exhibited dynamic morphological changes in the form of wave-like movements of the lamellipodium and dynamic protrusions on the platelet body. The increase in morphological activity was dependent on thrombin concentration. No increase in activity was observed following exposure to other activation agonists or during contact-induced activation. Inhibition of actin polymerization and inhibition of dynein significantly decreased the activity of thrombin-stimulated platelets. Our data suggest that these morphological dynamics after spreading are thrombin-specific and might play a role in coagulation and blood clot formation.
Collapse
Affiliation(s)
- Jan Seifert
- Institute of Applied Physics, University of Tübingen, Tübingen, Germany
| | | | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Cardiovascular Diseases, University of Tübingen, Tübingen, Germany
| | - Tilman E Schäffer
- Institute of Applied Physics, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
24
|
Abstract
MLCP (myosin light chain phosphatase) regulates platelet function through its ability to control myosin IIa phosphorylation. Recent evidence suggests that MLCP is a de facto target for signalling events stimulated by cAMP. In the present mini-review, we discuss the mechanisms by which cAMP signalling maintains MLCP in an active state to control platelet contractile machinery.
Collapse
|
25
|
Zhai K, Tang Y, Zhang Y, Li F, Wang Y, Cao Z, Yu J, Kou J, Yu B. NMMHC IIA inhibition impedes tissue factor expression and venous thrombosis via Akt/GSK3β-NF-κB signalling pathways in the endothelium. Thromb Haemost 2015; 114:173-85. [PMID: 25881103 DOI: 10.1160/th14-10-0880] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 02/19/2015] [Indexed: 01/29/2023]
Abstract
Non-muscle myosin heavy chain IIA (NMMHC IIA) has been shown to be involved in thrombus formation and inflammatory microparticle release in endothelial cells. However, the role of NMMHC IIA in regulating the expression of tissue factor (TF) and deep venous thrombosis remains to be elucidated. In the present study, endothelial cells were stimulated with tumour necrosis factor-α (TNF-α) to induce TF expression. Pretreatment with the NMMHC II inhibitor blebbistatin suppressed the mRNA and protein expressions as well as the procoagulant activity of TF in a dose-dependent manner. Blebbistatin enhanced Akt and GSK3β phosphorylation and inhibited NF-κB p65 nuclear translocation and IκBα degradation. These observations were similar to the effect of CHIR99021, a GSK3β inhibitor. TF downregulation by blebbistatin was antagonised by the PI3K inhibitor, wortmannin. Furthermore, siRNA knockdown of NMMHC IIA, but not IIB or IIC, inhibited TF expression, activated Akt/GSK3β and suppressed NF-κB signalling pathways, whereas the overexpression of NMMHC IIA increased TF expression. The binding of NMMHC IIA and TNF receptor 2 mediated signal internalisation in TNF-α-stimulated endothelial cells. Importantly, blebbistatin decreased endothelium NMMHC IIA and TF expression, deactivated GSK3β by inducing its phosphorylation, suppressed p65 nuclear translocation, and inhibited thrombus formation in a mouse deep venous thrombosis model.Our findings provide solid evidence that inhibition of NMMHC II, most likely NMMHC IIA, impedes TF expression and venous thrombosis via Akt/GSK3β-NF-κB signalling pathways in the endothelium both in vitro and in vivo. NMMHC IIA might be a potential novel target for the treatment of thrombotic disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jun Yu
- Dr. Jun Yu, Department of Internal Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT 06519, USA, Tel.: +1 203 7372869, Fax: +1 203 7372290, E-mail:
| | - Junping Kou
- Dr. Junping Kou, State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCM, China Pharmaceutical University, 639 Longmian Road, Nanjing, 211198, P. R. China, Tel./Fax: +86 25 86185158, E-mail:
| | | |
Collapse
|
26
|
A systems approach to hemostasis: 3. Thrombus consolidation regulates intrathrombus solute transport and local thrombin activity. Blood 2014; 124:1824-31. [PMID: 24951426 DOI: 10.1182/blood-2014-01-550319] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Hemostatic thrombi formed after a penetrating injury have a distinctive structure in which a core of highly activated, closely packed platelets is covered by a shell of less-activated, loosely packed platelets. We have shown that differences in intrathrombus molecular transport emerge in parallel with regional differences in platelet packing density and predicted that these differences affect thrombus growth and stability. Here we test that prediction in a mouse vascular injury model. The studies use a novel method for measuring thrombus contraction in vivo and a previously characterized mouse line with a defect in integrin αIIbβ3 outside-in signaling that affects clot retraction ex vivo. The results show that the mutant mice have a defect in thrombus consolidation following vascular injury, resulting in an increase in intrathrombus transport rates and, as predicted by computational modeling, a decrease in thrombin activity and platelet activation in the thrombus core. Collectively, these data (1) demonstrate that in addition to the activation state of individual platelets, the physical properties of the accumulated mass of adherent platelets is critical in determining intrathrombus agonist distribution and platelet activation and (2) define a novel role for integrin signaling in the regulation of intrathrombus transport rates and localization of thrombin activity.
Collapse
|
27
|
Menter DG, Tucker SC, Kopetz S, Sood AK, Crissman JD, Honn KV. Platelets and cancer: a casual or causal relationship: revisited. Cancer Metastasis Rev 2014; 33:231-69. [PMID: 24696047 PMCID: PMC4186918 DOI: 10.1007/s10555-014-9498-0] [Citation(s) in RCA: 227] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human platelets arise as subcellular fragments of megakaryocytes in bone marrow. The physiologic demand, presence of disease such as cancer, or drug effects can regulate the production circulating platelets. Platelet biology is essential to hemostasis, vascular integrity, angiogenesis, inflammation, innate immunity, wound healing, and cancer biology. The most critical biological platelet response is serving as "First Responders" during the wounding process. The exposure of extracellular matrix proteins and intracellular components occurs after wounding. Numerous platelet receptors recognize matrix proteins that trigger platelet activation, adhesion, aggregation, and stabilization. Once activated, platelets change shape and degranulate to release growth factors and bioactive lipids into the blood stream. This cyclic process recruits and aggregates platelets along with thrombogenesis. This process facilitates wound closure or can recognize circulating pathologic bodies. Cancer cell entry into the blood stream triggers platelet-mediated recognition and is amplified by cell surface receptors, cellular products, extracellular factors, and immune cells. In some cases, these interactions suppress immune recognition and elimination of cancer cells or promote arrest at the endothelium, or entrapment in the microvasculature, and survival. This supports survival and spread of cancer cells and the establishment of secondary lesions to serve as important targets for prevention and therapy.
Collapse
Affiliation(s)
- David G Menter
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | | | | | | | | | | |
Collapse
|
28
|
Diagouraga B, Grichine A, Fertin A, Wang J, Khochbin S, Sadoul K. Motor-driven marginal band coiling promotes cell shape change during platelet activation. ACTA ACUST UNITED AC 2014; 204:177-85. [PMID: 24421335 PMCID: PMC3897189 DOI: 10.1083/jcb.201306085] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
During platelet activation, motor protein-induced coiling of the microtubule-based marginal band leads to the cells’ characteristic spherical shape, whereas actomyosin-mediated compression of the coil results in new microtubule polymerization in a smaller ring. Platelets float in the blood as discoid particles. Their shape is maintained by microtubules organized in a ring structure, the so-called marginal band (MB), in the periphery of resting platelets. Platelets are activated after vessel injury and undergo a major shape change known as disc to sphere transition. It has been suggested that actomyosin tension induces the contraction of the MB to a smaller ring. In this paper, we show that antagonistic microtubule motors keep the MB in its resting state. During platelet activation, dynein slides microtubules apart, leading to MB extension rather than contraction. The MB then starts to coil, thereby inducing the spherical shape of activating platelets. Newly polymerizing microtubules within the coiled MB will then take a new path to form the smaller microtubule ring, in concerted action with actomyosin tension. These results present a new view of the platelet activation mechanism and reveal principal mechanistic features underlying cellular shape changes.
Collapse
Affiliation(s)
- Boubou Diagouraga
- Institut Albert Bonniot, Institut National de la Santé et de la Recherche Médicale U823; and 2 Techniques de l'Ingénierie Médicale et de la Complexité-Informatique, Mathématiques et Applications, Grenoble, Centre National de la Recherche Scientifique Unité Mixte de Recherche 5525; Université Joseph Fourier-Grenoble 1, F-38041 Grenoble, France
| | | | | | | | | | | |
Collapse
|
29
|
Cosemans JMEM, Angelillo-Scherrer A, Mattheij NJA, Heemskerk JWM. The effects of arterial flow on platelet activation, thrombus growth, and stabilization. Cardiovasc Res 2013; 99:342-52. [PMID: 23667186 DOI: 10.1093/cvr/cvt110] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Injury of an arterial vessel wall acutely triggers a multifaceted process of thrombus formation, which is dictated by the high-shear flow conditions in the artery. In this overview, we describe how the classical concept of arterial thrombus formation and vascular occlusion, driven by platelet activation and fibrin formation, can be extended and fine-tuned. This has become possible because of recent insight into the mechanisms of: (i) platelet-vessel wall and platelet-platelet communication, (ii) autocrine platelet activation, and (iii) platelet-coagulation interactions, in relation to blood flow dynamics. We list over 40 studies with genetically modified mice showing a role of platelet and plasma proteins in the control of thrombus stability after vascular injury. These include multiple platelet adhesive receptors and other junctional molecules, components of the ADP receptor signalling cascade to integrin activation, proteins controlling platelet shape, and autocrine activation processes, as well as multiple plasma proteins binding to platelets and proteins of the intrinsic coagulation cascade. Regulatory roles herein of the endothelium and other blood cells are recapitulated as well. Patient studies support the contribution of platelet- and coagulation activation in the regulation of thrombus stability. Analysis of the factors determining flow-dependent thrombus stabilization and embolus formation in mice will help to understand the regulation of this process in human arterial disease.
Collapse
Affiliation(s)
- Judith M E M Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht , Maastricht University, PO Box 616, Maastricht 6200 MD, The Netherlands
| | | | | | | |
Collapse
|
30
|
Abstract
The Rho family of GTP binding proteins, also commonly referred to as the Rho GTPases, are master regulators of the platelet cytoskeleton and platelet function. These low-molecular-weight or 'small' GTPases act as signaling switches in the spatial and temporal transduction, and amplification of signals from platelet cell surface receptors to the intracellular signaling pathways that drive platelet function. The Rho GTPase family members RhoA, Cdc42 and Rac1 have emerged as key regulators in the dynamics of the actin cytoskeleton in platelets and play key roles in platelet aggregation, secretion, spreading and thrombus formation. Rho GTPase regulators, including GEFs and GAPs and downstream effectors, such as the WASPs, formins and PAKs, may also regulate platelet activation and function. In this review, we provide an overview of Rho GTPase signaling in platelet physiology. Previous studies of Rho GTPases and platelets have had a shared history, as platelets have served as an ideal, non-transformed cellular model to characterize Rho function. Likewise, recent studies of the cell biology of Rho GTPase family members have helped to build an understanding of the molecular regulation of platelet function and will continue to do so through the further characterization of Rho GTPases as well as Rho GAPs, GEFs, RhoGDIs and Rho effectors in actin reorganization and other Rho-driven cellular processes.
Collapse
Affiliation(s)
- J E Aslan
- Department of Biomedical Engineering and Cell & Developmental Biology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA.
| | | |
Collapse
|
31
|
Schwarz Henriques S, Sandmann R, Strate A, Köster S. Force field evolution during human blood platelet activation. J Cell Sci 2012; 125:3914-20. [PMID: 22582082 DOI: 10.1242/jcs.108126] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Contraction at the cellular level is vital for living organisms. The most prominent type of contractile cells are heart muscle cells, a less-well-known example is blood platelets. Blood platelets activate and interlink at injured blood vessel sites, finally contracting to form a compact blood clot. They are ideal model cells to study the mechanisms of cellular contraction, as they are simple, having no nucleus, and their activation can be triggered and synchronized by the addition of thrombin. We have studied contraction using human blood platelets, employing traction force microscopy, a single-cell technique that enables time-resolved measurements of cellular forces on soft substrates with elasticities in the physiological range (∼4 kPa). We found that platelet contraction reaches a steady state after 25 min with total forces of ∼34 nN. These forces are considerably larger than what was previously reported for platelets in aggregates, demonstrating the importance of a single-cell approach for studies of platelet contraction. Compared with other contractile cells, we find that platelets are unique, because force fields are nearly isotropic, with forces pointing toward the center of the cell area.
Collapse
Affiliation(s)
- Sarah Schwarz Henriques
- University of Göttingen, Department of X-Ray Physics and Courant Research Centre Nano-Spectroscopy and X-Ray Imaging, 37077 Göttingen, Germany
| | | | | | | |
Collapse
|
32
|
Abstract
Hemostasis is dependent upon the successful recruitment and activation of blood platelets to the site of a breach in the vasculature. Platelet activation stimulates the rapid reorganization of the cortical actin cytoskeleton, resulting in the transformation of platelets from biconcave disks to fully spread cells. During this process, platelets extend filopodia and generate lamellipodia, resulting in a dramatic increase in the platelet surface area. Kohler-illuminated Nomarski Differential Interference Contrast microscopy has proved an effective tool to characterize platelet morphological changes in real time, and provides a useful tool to identify genetic and pharmacological regulators of platelet function.
Collapse
|
33
|
Maravillas-Montero JL, Santos-Argumedo L. The myosin family: unconventional roles of actin-dependent molecular motors in immune cells. J Leukoc Biol 2011; 91:35-46. [DOI: 10.1189/jlb.0711335] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
34
|
Evidence for involvement of ROCK signaling in bradykinin-induced increase in murine blood–tumor barrier permeability. J Neurooncol 2011; 106:291-301. [DOI: 10.1007/s11060-011-0685-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Accepted: 08/03/2011] [Indexed: 01/23/2023]
|
35
|
Thomas SG, Calaminus SDJ, Machesky LM, Alberts AS, Watson SP. G-protein coupled and ITAM receptor regulation of the formin FHOD1 through Rho kinase in platelets. J Thromb Haemost 2011; 9:1648-51. [PMID: 21605332 DOI: 10.1111/j.1538-7836.2011.04357.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
36
|
Brass LF, Wannemacher KM, Ma P, Stalker TJ. Regulating thrombus growth and stability to achieve an optimal response to injury. J Thromb Haemost 2011; 9 Suppl 1:66-75. [PMID: 21781243 PMCID: PMC3422128 DOI: 10.1111/j.1538-7836.2011.04364.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
An optimal platelet response to injury can be defined as one in which blood loss is restrained and haemostasis is achieved without the penalty of further tissue damage caused by unwarranted vascular occlusion. This brief review considers some of the ways in which thrombus growth and stability can be regulated so that an optimal platelet response can be achieved in vivo. Three related topics are considered. The first focuses on intracellular mechanisms that regulate the early events of platelet activation downstream of G protein coupled receptors for agonists such as thrombin, thromboxane A(2) and ADP. The second considers the ways in which signalling events that are dependent on stable contacts between platelets can influence the state of platelet activation and thus affect thrombus growth and stability. The third focuses on the changes that are experienced by platelets as they move from their normal environment in freely-flowing plasma to a very different environment within the growing haemostatic plug, an environment in which the narrowing gaps and junctions between platelets not only facilitate communication, but also increasingly limit both the penetration of plasma and the exodus of platelet-derived bioactive molecules.
Collapse
Affiliation(s)
- L F Brass
- Department of Medicine and Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
37
|
Lasne D, Baujat G, Mirault T, Lunardi J, Grelac F, Egot M, Salomon R, Bachelot-Loza C. Bleeding disorders in Lowe syndrome patients: evidence for a link between OCRL mutations and primary haemostasis disorders. Br J Haematol 2010; 150:685-8. [PMID: 20629659 DOI: 10.1111/j.1365-2141.2010.08304.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Lowe syndrome (LS) is a rare X-linked disorder caused by mutations in the oculocerebrorenal gene (OCRL), encoding OCRL, a phosphatidylinositol 5-phosphatase with a RhoGAP domain. An abnormal rate of haemorrhagic events was found in a retrospective clinical survey. Herein, we report the results of exploration of haemostasis in six LS patients. All patients had normal coagulation tests but prolonged closure times (CTs) in the PFA-100 system. Healthy donors' blood samples incubated with a RhoA kinase inhibitor had prolonged CTs. This suggests that an aberrant RhoA pathway in platelets contributes to CT prolongation and primary haemostasis disorders in LS.
Collapse
Affiliation(s)
- Dominique Lasne
- AP-HP Hôpital Necker, Laboratoire d'Hématologie, Paris, France.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Catricalà S, Guidetti GF, Canobbio I, Pecci A, Balduini CL, Balduini C, Torti M. The irreversibility of platelet aggregation is regulated by myosin IIA, but is not compromised in MYH9-related disease. Thromb Res 2010; 127:171-3. [PMID: 20828798 DOI: 10.1016/j.thromres.2010.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 08/03/2010] [Accepted: 08/04/2010] [Indexed: 10/19/2022]
|
39
|
Liang XM, Han SJ, Reems JA, Gao DY, Sniadecki NJ. Platelet retraction force measurements using flexible post force sensors. LAB ON A CHIP 2010; 10:991-8. [PMID: 20358105 PMCID: PMC4918627 DOI: 10.1039/b918719g] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Platelets play an important role in hemostasis by forming a thrombotic plug that seals the vessel wall and promotes vascular healing. After platelets adhere and aggregate at the wound site, their next step is to generate contractile forces through the coordination of physicochemical interactions between actin, myosin, and alpha(IIb)beta(3) integrin receptors that retract the thrombus' size and strengthen its adhesion to the exposed matrix. Although platelet contractile forces (PCF) are a definitive feature of hemostasis and thrombosis, there are few approaches that can directly measure them. In this study, we describe the development of an approach to measure PCF in microthrombi using a microscopic flexible post force sensor array. Quasi-static measurements and live microscopic imaging of thrombin-activated platelets on the posts were conducted to assay the development of PCF to various hemostatic conditions. Microthrombi were observed to produce forces that monotonically increased with thrombin concentration and activation time, but forces subsided when thrombin was removed. PCF results were statistically similar on arrays of posts printed with fibronectin or fibrinogen. PCF measurements were combined with clot volume measurements to determine that the average force per platelet was 2.1 +/- 0.1 nN after 60 min, which is significantly higher than what has been measured with previous approaches. Overall, the flexible post arrays for PCF measurements are a promising approach for evaluating platelet functionality, platelet physiology and pathology, the impacts of different matrices or agonists on hemostatic responses, and in providing critical information regarding platelet activity that can guide new hemostatic or thrombotic strategies.
Collapse
Affiliation(s)
- Xin M. Liang
- Department of Mechanical Engineering, University of Washington, Seattle, USA
| | - Sangyoon J. Han
- Department of Mechanical Engineering, University of Washington, Seattle, USA
| | | | - Dayong Y. Gao
- Department of Mechanical Engineering, University of Washington, Seattle, USA
| | - Nathan J. Sniadecki
- Department of Mechanical Engineering, University of Washington, Seattle, USA
| |
Collapse
|
40
|
Tournoij E, Weber GJ, Akkerman JWN, de Groot PG, Zon LI, Moll FL, Schulte-Merker S. Mlck1a is expressed in zebrafish thrombocytes and is an essential component of thrombus formation. J Thromb Haemost 2010; 8:588-95. [PMID: 20002541 PMCID: PMC2935642 DOI: 10.1111/j.1538-7836.2009.03721.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND We have used the advantages of the zebrafish model system to demonstrate which of the vertebrate myosin light chain kinase (MLCK) genes is expressed in thrombocytes and important for thrombus formation. METHODS AND RESULTS Here we report that Mlck1a is an essential component of thrombus formation. Phylogenetic data revealed four zebrafish orthologous for three human MLCK genes. To investigate expression of the zebrafish mlck genes in thrombocytes we compared GFP-tagged platelets with other cells by microarray analysis, and showed that mlck1a expression was 4.5-fold enriched in platelets. Furthermore, mlck1a mRNA and mRNA for the platelet-specific cd41 co-localized in thrombi. Expression of other mlck subtypes was lower in GFP-tagged platelets (mlck1b; 0.77-fold enriched) and absent in thrombi (mlck1b, -2, -3). To investigate the role of Mlck1a in thrombus formation, we knocked down mlck1a using two morpholinos. This resulted in impaired morphology changes of platelets adhering on fibrinogen. In a thrombosis model, in which thrombocytes adhere to the vessel wall damaged by laser irradiation, thrombus formation was slowed down in mlck1a-deficient embryos. CONCLUSION We conclude that Mlck1a is the subtype of MLCK that contributes to platelet shape change and thrombus formation.
Collapse
Affiliation(s)
- E Tournoij
- Hubrecht Institute-KNAW and UMC, Utrecht, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
41
|
Cai Y, Rossier O, Gauthier NC, Biais N, Fardin MA, Zhang X, Miller LW, Ladoux B, Cornish VW, Sheetz MP. Cytoskeletal coherence requires myosin-IIA contractility. J Cell Sci 2010; 123:413-23. [PMID: 20067993 DOI: 10.1242/jcs.058297] [Citation(s) in RCA: 156] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Maintaining a physical connection across cytoplasm is crucial for many biological processes such as matrix force generation, cell motility, cell shape and tissue development. However, in the absence of stress fibers, the coherent structure that transmits force across the cytoplasm is not understood. We find that nonmuscle myosin-II (NMII) contraction of cytoplasmic actin filaments establishes a coherent cytoskeletal network irrespective of the nature of adhesive contacts. When NMII activity is inhibited during cell spreading by Rho kinase inhibition, blebbistatin, caldesmon overexpression or NMIIA RNAi, the symmetric traction forces are lost and cell spreading persists, causing cytoplasm fragmentation by membrane tension that results in 'C' or dendritic shapes. Moreover, local inactivation of NMII by chromophore-assisted laser inactivation causes local loss of coherence. Actin filament polymerization is also required for cytoplasmic coherence, but microtubules and intermediate filaments are dispensable. Loss of cytoplasmic coherence is accompanied by loss of circumferential actin bundles. We suggest that NMIIA creates a coherent actin network through the formation of circumferential actin bundles that mechanically link elements of the peripheral actin cytoskeleton where much of the force is generated during spreading.
Collapse
Affiliation(s)
- Yunfei Cai
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Liu Z, van Grunsven LA, Van Rossen E, Schroyen B, Timmermans JP, Geerts A, Reynaert H. Blebbistatin inhibits contraction and accelerates migration in mouse hepatic stellate cells. Br J Pharmacol 2009; 159:304-15. [PMID: 20039876 DOI: 10.1111/j.1476-5381.2009.00477.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Blebbistatin, an inhibitor of myosin-II-specific ATPase, has been used to inhibit contraction of invertebrate and mammalian muscle preparations containing non-muscle myosin. Activated hepatic stellate cells have contractile properties and play an important role in the pathophysiology of liver fibrosis and portal hypertension. Therefore, hepatic stellate cells are considered as therapeutic target cells. In the present study, we studied the effect of blebbistatin during the transition of mouse hepatic stellate cells into contractile myofibroblasts. EXPERIMENTAL APPROACH Effects of blebbistatin on cell morphology were evaluated by phase contrast microscopy. Cell stress fibres and focal adhesions were investigated by dual immunofluorescence staining and visualized using fluorescence microscopy. Contractile force generation was examined by silicone wrinkle formation assays and collagen gel contraction assays. Intracellular Ca(2+) release in response to endothelin-1 was measured by using Fluo-4. Cell migration was measured by wound healing experiments. KEY RESULTS In culture-activated hepatic stellate cells, blebbistatin was found to change both cell morphology and function. In the presence of blebbistatin, stellate cells became smaller, acquired a dendritic morphology and had less myosin IIA-containing stress fibres and vinculin-containing focal adhesions. Moreover, blebbistatin impaired silicone wrinkle formation, reduced collagen gel contraction and blocked endothelin-1-induced intracellular Ca(2+) release. Finally, it promoted wound-induced cell migration. CONCLUSIONS AND IMPLICATIONS By inhibiting myosin II ATPase, blebbistatin has profound effects on the morphology and function of activated hepatic stellate cells. Our data suggest that myosin II could be a therapeutic target in the treatment of liver fibrosis and portal hypertension.
Collapse
Affiliation(s)
- Zhenan Liu
- Department of Cell Biology, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|
43
|
Non-muscle myosin II takes centre stage in cell adhesion and migration. Nat Rev Mol Cell Biol 2009; 10:778-90. [PMID: 19851336 DOI: 10.1038/nrm2786] [Citation(s) in RCA: 1401] [Impact Index Per Article: 93.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Non-muscle myosin II (NM II) is an actin-binding protein that has actin cross-linking and contractile properties and is regulated by the phosphorylation of its light and heavy chains. The three mammalian NM II isoforms have both overlapping and unique properties. Owing to its position downstream of convergent signalling pathways, NM II is central in the control of cell adhesion, cell migration and tissue architecture. Recent insight into the role of NM II in these processes has been gained from loss-of-function and mutant approaches, methods that quantitatively measure actin and adhesion dynamics and the discovery of NM II mutations that cause monogenic diseases.
Collapse
|
44
|
Laschke MW, Dold S, Menger MD, Jeppsson B, Thorlacius H. The Rho-kinase inhibitor Y-27632 inhibits cholestasis-induced platelet interactions in the hepatic microcirculation. Microvasc Res 2009; 78:95-9. [PMID: 19374910 DOI: 10.1016/j.mvr.2009.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Revised: 03/06/2009] [Accepted: 04/07/2009] [Indexed: 10/20/2022]
Abstract
Bile duct obstruction is associated with hepatic accumulation of leukocytes and liver injury. Emerging data suggest that platelets may play an important role in tissue damage and inflammation. Herein, we characterized the platelet response in cholestatic liver injury and evaluated the role of Rho-kinase signaling. For this purpose, C57BL/6 mice were treated with the Rho-kinase inhibitor Y-27632 (10 mg/kg) and vehicle before undergoing bile duct ligation (BDL) for 12 h. Platelet rolling and adhesion, formation of platelet aggregates as well as microvascular perfusion in the liver were analyzed using intravital fluorescence microscopy. Liver damage was monitored by measuring serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST). Administration of Y-27632 reduced the BDL-associated increase of ALT and AST by 95% and 89%, respectively. The inhibition of Rho-kinase also reduced cholestasis-induced platelet rolling and adhesion by more than 46% and 73% in postsinusoidal venules and platelet adhesion in sinusoids by 60%. In addition, Y-27632 decreased platelet aggregation in hepatic sinusoids and postsinusoidal venules by 69% and 81%. BDL caused a significant reduction of hepatic microvascular perfusion. Importantly, pretreatment with Y-27632 restored sinusoidal perfusion in cholestatic animals. Our findings demonstrate that Rho-kinase regulates multiple aspects of platelet interaction in the microcirculation of cholestatic animals. Moreover, inhibition of Rho-kinase signaling not only attenuates platelet responses but also maintains microvascular perfusion and protects against hepatocellular injury in cholestasis. Thus, targeting Rho-kinase signaling may be an effective way to protect against platelet-mediated liver injury in obstructive jaundice.
Collapse
Affiliation(s)
- Matthias W Laschke
- Department of Surgery, Malmö University Hospital, Malmö, Lund University, Sweden.
| | | | | | | | | |
Collapse
|
45
|
Calaminus SDJ, Thomas S, McCarty OJT, Machesky LM, Watson SP. Identification of a novel, actin-rich structure, the actin nodule, in the early stages of platelet spreading. J Thromb Haemost 2008; 6:1944-52. [PMID: 18761725 DOI: 10.1111/j.1538-7836.2008.03141.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND During platelet spreading, the actin cytoskeleton undergoes marked changes, forming filopodia, lamellipodia and stress fibres. In the present study, we report the identification of a novel actin-rich structure, termed an actin nodule, which appears prior to lamellipodia and stress fibre formation. METHODS Platelet spreading was monitored using human platelets and mouse GFP-actin platelets using real-time and end-point DIC, and fluorescent and electron microscopy (EM). RESULTS We identified a small, novel actin structure, the actin nodule, in the early stages of adhesion and spreading, which we hypothesize to be a precursor of lamellipodia and stress fibres. Nodule formation shows an inverse correlation to Rho kinase and myosin-II activity, is independent of PI3-kinase, but dependent on Src kinase activity. Actin nodules contain multiple proteins, including Arp2/3, Fyn, Rac, and beta1- and beta3- integrins, but not Src. EM analysis revealed that actin filaments extend in all directions from the nodules. Actin nodules are present on multiple matrices, including fibrinogen, laminin and VWF + botrocetin. CONCLUSION This work identifies a novel platelet actin structure, which we propose is a precursor to both lamellipodia and stress fibres and acts to drive platelet spreading.
Collapse
Affiliation(s)
- S D J Calaminus
- Centre for Cardiovascular Sciences, Institute of Biomedical Research, The Medical School, University of Birmingham, Birmingham, UK.
| | | | | | | | | |
Collapse
|
46
|
Auger JM, Watson SP. Dynamic Tyrosine Kinase-Regulated Signaling and Actin Polymerisation Mediate Aggregate Stability Under Shear. Arterioscler Thromb Vasc Biol 2008; 28:1499-504. [DOI: 10.1161/atvbaha.108.167296] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Objective—
Aggregate formation on collagen at arteriolar rates of shear is mediated by coordinated signaling between tyrosine kinase–linked and G protein–coupled receptors. We have investigated the role of these receptors and the actin cytoskeleton in maintaining aggregate stability under shear.
Methods and Results—
Platelet aggregates are rapidly formed when blood is flowed over collagen at 1000 s
−1
and remain stable over 20 minutes. A novel fibrin-independent mechanism of retraction against the direction of flow occurs at the aggregate front and recruits platelets into the main aggregate. Stable aggregates are not observed in the presence of cytochalasin D, which blocks de novo actin polymerization. When exposed to the Src family kinase inhibitor, PD0173952, preformed aggregates spread in the direction of flow and rounded platelets appear within the aggregate body and are lost in the direction of flow. A similar set of observations is observed in the presence of latrunculin A, which disrupts preexisting actin filaments, but not in the combined presence of inhibitors of ADP and thromboxane A
2
formation.
Conclusions—
Maintenance of stable aggregates at high shear is a dynamic process mediated by Src kinases and actin polymerization. These signals maintain aggregates in a compact structure and prevent continuous streaming of platelets.
Collapse
Affiliation(s)
- Jocelyn M. Auger
- From the Centre for Cardiovascular Sciences, Institute of Biomedical Research, The Medical School, University of Birmingham, UK
| | - Steve P. Watson
- From the Centre for Cardiovascular Sciences, Institute of Biomedical Research, The Medical School, University of Birmingham, UK
| |
Collapse
|
47
|
Identification of a fibrin-independent platelet contractile mechanism regulating primary hemostasis and thrombus growth. Blood 2008; 112:90-9. [DOI: 10.1182/blood-2007-12-127001] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractA fundamental property of platelets is their ability to transmit cytoskeletal contractile forces to extracellular matrices. While the importance of the platelet contractile mechanism in regulating fibrin clot retraction is well established, its role in regulating the primary hemostatic response, independent of blood coagulation, remains ill defined. Real-time analysis of platelet adhesion and aggregation on a collagen substrate revealed a prominent contractile phase during thrombus development, associated with a 30% to 40% reduction in thrombus volume. Thrombus contraction developed independent of thrombin and fibrin and resulted in the tight packing of aggregated platelets. Inhibition of the platelet contractile mechanism, with the myosin IIA inhibitor blebbistatin or through Rho kinase antagonism, markedly inhibited thrombus contraction, preventing the tight packing of aggregated platelets and undermining thrombus stability in vitro. Using a new intravital hemostatic model, we demonstrate that the platelet contractile mechanism is critical for maintaining the integrity of the primary hemostatic plug, independent of thrombin and fibrin generation. These studies demonstrate an important role for the platelet contractile mechanism in regulating primary hemostasis and thrombus growth. Furthermore, they provide new insight into the underlying bleeding diathesis associated with platelet contractility defects.
Collapse
|