1
|
Maulana TI, Wevers NR, Kristoforus T, Chandler M, Lanz HL, Joore J, Vulto P, Villenave R, Kustermann S, Loskill P, Bircsak KM. Opportunities for Microphysiological Systems in Toxicity Testing of New Drug Modalities. Annu Rev Pharmacol Toxicol 2025; 65:47-69. [PMID: 39227343 DOI: 10.1146/annurev-pharmtox-061724-080621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
New drug modalities offer life-saving benefits for patients through access to previously undruggable targets. Yet these modalities pose a challenge for the pharmaceutical industry, as side effects are complex, unpredictable, and often uniquely human. With animal studies having limited predictive value due to translatability challenges, the pharmaceutical industry seeks out new approach methodologies. Microphysiological systems (MPS) offer important features that enable complex toxicological processes to be modeled in vitro such as (a) an adjustable complexity of cellular components, including immune components; (b) a modifiable tissue architecture; (c) integration and monitoring of dynamic mechanisms; and (d) a multiorgan connection. Here we review MPS studies in the context of four clinical adverse events triggered by new drug modalities: peripheral neuropathy, thrombocytopenia, immune-mediated hepatotoxicity, and cytokine release syndrome. We conclude that while the use of MPS for testing new drug modality-induced toxicities is still in its infancy, we see strong potential going forward.
Collapse
Affiliation(s)
- Tengku Ibrahim Maulana
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | | | - Theodora Kristoforus
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | | | | | - Jos Joore
- MIMETAS BV, Oegstgeest, The Netherlands
| | | | - Remi Villenave
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Stefan Kustermann
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Peter Loskill
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | | |
Collapse
|
2
|
Ranjbar J, Yang Y, Harper AGS. Developing human tissue engineered arterial constructs to simulate human in vivo thrombus formation. Platelets 2023; 34:2153823. [PMID: 36550074 DOI: 10.1080/09537104.2022.2153823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Thrombus formation is highly dependent upon the physico-chemical environment in which it is triggered. Our ability to understand how thrombus formation is initiated, regulated, and resolved in the human body is dependent upon our ability to replicate the mechanical and biological properties of the arterial wall. Current in vitro thrombosis models principally use reductionist approaches to model the complex biochemical and cellular milieu present in the arterial wall, and so researcher have favored the use of in vivo models. The field of vascular tissue engineering has developed a range of techniques for culturing artificial human arteries for use as vascular grafts. These techniques therefore provide a basis for developing more sophisticated 3D replicas of the arterial wall that can be used in in vitro thrombosis models. In this review, we consider how tissue engineering approaches can be used to generate 3D models of the arterial wall that improve upon current in vivo and in vitro approaches. We consider the current benefits and limitations of reported 3D tissue engineered models and consider what additional evidence is required to validate them as alternatives to current in vivo models.
Collapse
Affiliation(s)
| | - Ying Yang
- School of Pharmacy & Bioengineering, Keele University, Keele, UK
| | | |
Collapse
|
3
|
Sakurai Y, Hardy ET, Lam WA. Hemostasis-on-a-chip / incorporating the endothelium in microfluidic models of bleeding. Platelets 2023; 34:2185453. [PMID: 36872890 PMCID: PMC10197822 DOI: 10.1080/09537104.2023.2185453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 02/23/2023] [Indexed: 03/07/2023]
Abstract
Currently, point-of-care assays for human platelet function and coagulation are used to assess bleeding risks and drug testing, but they lack intact endothelium, a critical component of the human vascular system. Within these assays, the assessment of bleeding risk is typically indicated by the lack of or reduced platelet function and coagulation without true evaluation of hemostasis. Hemostasis is defined as the cessation of bleeding. Additionally, animal models of hemostasis also, by definition, lack human endothelium, which may limit their clinical relevance. This review discusses the current state-of-the-art of hemostasis-on-a-chip, specifically, human cell-based microfluidic models that incorporate endothelial cells, which function as physiologically relevant in vitro models of bleeding. These assays recapitulate the entire process of vascular injury, bleeding, and hemostasis, and provide real-time, direct observation, thereby serving as research-enabling tools that enhance our understanding of hemostasis and also as novel drug discovery platforms.
Collapse
Affiliation(s)
- Yumiko Sakurai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Center of Children’s Healthcare of Atlanta, Emory University School of Medicine, Atlanta, USA
| | - Elaissa T. Hardy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Center of Children’s Healthcare of Atlanta, Emory University School of Medicine, Atlanta, USA
| | - Wilbur A. Lam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Center of Children’s Healthcare of Atlanta, Emory University School of Medicine, Atlanta, USA
| |
Collapse
|
4
|
Abstract
This review discusses our understanding of platelet diversity with implications for the roles of platelets in hemostasis and thrombosis and identifies advanced technologies set to provide new insights. We use the term diversity to capture intrasubject platelet variability that can be intrinsic or governed by the environment and lead to a heterogeneous response pattern of aggregation, clot promotion, and external communication. Using choice examples, we discuss how the use of advanced technologies can provide new insights into the underlying causes of platelet molecular, structural, and functional diversity. As sources of diversity, we discuss the proliferating megakaryocytes with different allele-specific expression patterns, the asymmetrical formation of proplatelets, changes in platelets induced by aging and priming, interplatelet heterogeneity in thrombus organization and stability, and platelet-dependent communications. We provide indications how current knowledge gaps can be addressed using promising technologies, such as next-generation sequencing, proteomic approaches, advanced imaging techniques, multicolor flow and mass cytometry, multifunctional microfluidics assays, and organ-on-a-chip platforms. We then argue how this technology base can aid in characterizing platelet populations and in identifying platelet biomarkers relevant for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands (J.W.M.H.)
| | - Jonathan West
- Faculty of Medicine and Centre for Hybrid Biodevices, University of Southampton, United Kingdom (J.W.)
| |
Collapse
|
5
|
Peters LJF, Baaten CCFMJ, Maas SL, Lu C, Nagy M, Jooss NJ, Bidzhekov K, Santovito D, Moreno-Andrés D, Jankowski J, Biessen EAL, Döring Y, Heemskerk JWM, Weber C, Kuijpers MJE, van der Vorst EPC. MicroRNA-26b Attenuates Platelet Adhesion and Aggregation in Mice. Biomedicines 2022; 10:983. [PMID: 35625720 PMCID: PMC9138361 DOI: 10.3390/biomedicines10050983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 02/05/2023] Open
Abstract
Platelets are key regulators of haemostasis, making platelet dysfunction a major driver of thrombosis. Numerous processes that determine platelet function are influenced by microRNAs (miRs). MiR-26b is one of the highest-expressed miRs in healthy platelets, and its expression in platelets is changed in a diseased state. However, the exact effect of this miR on platelet function has not been studied yet. In this study, we made use of a whole-body knockout of miR-26b in ApoE-deficient mice in order to determine its impact on platelet function, thrombus formation and platelet signalling both ex vivo and in vivo. We show that a whole-body deficiency of miR-26b exacerbated platelet adhesion and aggregation ex vivo. Additionally, in vivo, platelets adhered faster, and larger thrombi were formed in mice lacking miR-26b. Moreover, isolated platelets from miR-26b-deficient mice showed a hyperactivated Src and EGFR signalling. Taken together, we show here for the first time that miR-26b attenuates platelet adhesion and aggregation, possibly through Src and EGFR signalling.
Collapse
Affiliation(s)
- Linsey J. F. Peters
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52056 Aachen, Germany; (L.J.F.P.); (C.C.F.M.J.B.); (S.L.M.); (J.J.); (E.A.L.B.)
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52056 Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 Maastricht, The Netherlands;
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, 80337 Munich, Germany; (K.B.); (D.S.); (Y.D.); (C.W.)
| | - Constance C. F. M. J. Baaten
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52056 Aachen, Germany; (L.J.F.P.); (C.C.F.M.J.B.); (S.L.M.); (J.J.); (E.A.L.B.)
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 Maastricht, The Netherlands; (M.N.); (N.J.J.); (J.W.M.H.); (M.J.E.K.)
| | - Sanne L. Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52056 Aachen, Germany; (L.J.F.P.); (C.C.F.M.J.B.); (S.L.M.); (J.J.); (E.A.L.B.)
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52056 Aachen, Germany
| | - Chang Lu
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 Maastricht, The Netherlands;
| | - Magdolna Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 Maastricht, The Netherlands; (M.N.); (N.J.J.); (J.W.M.H.); (M.J.E.K.)
| | - Natalie J. Jooss
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 Maastricht, The Netherlands; (M.N.); (N.J.J.); (J.W.M.H.); (M.J.E.K.)
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Kiril Bidzhekov
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, 80337 Munich, Germany; (K.B.); (D.S.); (Y.D.); (C.W.)
| | - Donato Santovito
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, 80337 Munich, Germany; (K.B.); (D.S.); (Y.D.); (C.W.)
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, 80337 Munich, Germany
- Institute for Genetic and Biomedical Research (IRGB), Unit of Milan, National Research Council, 20090 Milan, Italy
| | - Daniel Moreno-Andrés
- Department of Biochemistry and Molecular Cell Biology, Medical School, RWTH Aachen University, 52056 Aachen, Germany;
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52056 Aachen, Germany; (L.J.F.P.); (C.C.F.M.J.B.); (S.L.M.); (J.J.); (E.A.L.B.)
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 Maastricht, The Netherlands;
| | - Erik A. L. Biessen
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52056 Aachen, Germany; (L.J.F.P.); (C.C.F.M.J.B.); (S.L.M.); (J.J.); (E.A.L.B.)
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 Maastricht, The Netherlands;
| | - Yvonne Döring
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, 80337 Munich, Germany; (K.B.); (D.S.); (Y.D.); (C.W.)
- Institute for Genetic and Biomedical Research (IRGB), Unit of Milan, National Research Council, 20090 Milan, Italy
- Swiss Cardiovascular Center, Division of Angiology, Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland
| | - Johan W. M. Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 Maastricht, The Netherlands; (M.N.); (N.J.J.); (J.W.M.H.); (M.J.E.K.)
- Synapse Research Institute, Kon. Emmaplein 7, 6217 Maastricht, The Netherlands
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, 80337 Munich, Germany; (K.B.); (D.S.); (Y.D.); (C.W.)
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 Maastricht, The Netherlands; (M.N.); (N.J.J.); (J.W.M.H.); (M.J.E.K.)
- Institute for Genetic and Biomedical Research (IRGB), Unit of Milan, National Research Council, 20090 Milan, Italy
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Marijke J. E. Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 Maastricht, The Netherlands; (M.N.); (N.J.J.); (J.W.M.H.); (M.J.E.K.)
- Thrombosis Expertise Center, Heart and Vascular Center, Maastricht University Medical Center, 6229 Maastricht, The Netherlands
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52056 Aachen, Germany; (L.J.F.P.); (C.C.F.M.J.B.); (S.L.M.); (J.J.); (E.A.L.B.)
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52056 Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 Maastricht, The Netherlands;
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, 80337 Munich, Germany; (K.B.); (D.S.); (Y.D.); (C.W.)
| |
Collapse
|
6
|
Forstner D, Guettler J, Gauster M. Changes in Maternal Platelet Physiology during Gestation and Their Interaction with Trophoblasts. Int J Mol Sci 2021; 22:10732. [PMID: 34639070 PMCID: PMC8509324 DOI: 10.3390/ijms221910732] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 01/06/2023] Open
Abstract
Upon activation, maternal platelets provide a source of proinflammatory mediators in the intervillous space of the placenta. Therefore, platelet-derived factors may interfere with different trophoblast subtypes of the developing human placenta and might cause altered hormone secretion and placental dysfunction later on in pregnancy. Increased platelet activation, and the subsequent occurrence of placental fibrinoid deposition, are linked to placenta pathologies such as preeclampsia. The composition and release of platelet-derived factors change over gestation and provide a potential source of predicting biomarkers for the developing fetus and the mother. This review indicates possible mechanisms of platelet-trophoblast interactions and discusses the effect of increased platelet activation on placenta development.
Collapse
Affiliation(s)
- Désirée Forstner
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (J.G.); (M.G.)
| | | | | |
Collapse
|
7
|
Ruane-O'Hora T, Markos F. Platelets Do Not Alter Flow-Mediated Dilation or Arterial Conduction in vivo. J Vasc Res 2021; 58:231-236. [PMID: 34010839 DOI: 10.1159/000516045] [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: 02/12/2021] [Accepted: 03/22/2021] [Indexed: 11/19/2022] Open
Abstract
The aim of this study was to investigate whether platelets contribute to shear stress and vascular conductance in the iliac vascular bed in vivo. Flow-mediated dilation of pig iliac was induced by downstream injection of acetylcholine (50 μg), and separately, conductance (ΔF/ΔP) was calculated. This was carried out before and after removal of 1 L of arterial blood in 240 mL increments, and each 240 mL was spun in a centrifuge (1,500 rcf for 7 min); platelet-rich plasma was replaced with equal volume of heparinised saline and reinjected. The circulating platelet count fell from 369 × 109/L (n = 5) to 165 × 109/L (p = 0.01; n = 4; Student's unpaired t). An increase in flow led to an increase in the iliac diameter by 0.49 ± 0.03 mm (mean ± SEM) before platelet reduction and 0.55 ± 0.05 mm after (p = 0.36, Student's paired t, n = 5); the change in arterial conductance was also not significantly affected by platelet reduction, control: 1.44 ± 0.34 mL/min/mm Hg, after platelet reduction: 1.39 ± 0.04 mm (p = 0.55, Student's paired t, n = 4). Therefore, platelets do not contribute to shear stress or conductance in vivo.
Collapse
Affiliation(s)
- Therese Ruane-O'Hora
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Farouk Markos
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| |
Collapse
|
8
|
von Willebrand factor variant D1472H has no effect in mice with humanized VWF-platelet interactions. Blood Adv 2021; 4:4065-4068. [PMID: 32870970 DOI: 10.1182/bloodadvances.2020002629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/31/2020] [Indexed: 01/06/2023] Open
Abstract
The von Willebrand factor ristocetin cofactor activity assay (VWF:RCo) is used for diagnosis of von Willebrand disease (VWD) because of its ability to evaluate VWF binding to platelets. VWF sequence variant p.D1472H is associated with lower VWF:RCo levels in the absence of associated bleeding symptoms, indicating the VWF:RCo may not be accurate for characterizing VWF function in individuals with this variant. Thus, this study aimed to determine the implications of the variant on VWF functioning in vivo. Mice were engineered with humanized wild-type (WT*) VWF A1/A2 and VWF with the p.D1472H (1472H) variant along with humanized platelet GPIbα and bred to homozygosity. VWF antigen and VWF binding to GPIbα were measured using enzyme-linked immunosorbent assay. Gel electrophoresis was used for VWF multimer analysis. Tail bleeding assays were performed at a 3-mm defined length. Normal VWF multimers were preserved in both WT* and 1472H mice. VWF expression was normal in the WT* and 1472H mice, and VWF binding to GPIbα did not statistically differ between the groups. Additionally, tail bleeding times were similar for WT* and 1472H mice. These results show the p.D1472H variant does not impair hemostasis in mice, and support the conclusion that p.D1472H is a normal variant in humans.
Collapse
|
9
|
Poventud-Fuentes I, Kwon KW, Seo J, Tomaiuolo M, Stalker TJ, Brass LF, Huh D. A Human Vascular Injury-on-a-Chip Model of Hemostasis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2004889. [PMID: 33150735 PMCID: PMC8049960 DOI: 10.1002/smll.202004889] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/28/2020] [Indexed: 05/02/2023]
Abstract
Hemostasis is an innate protective mechanism that plays a central role in maintaining the homeostasis of the vascular system during vascular injury. Studying this essential physiological process is often challenged by the difficulty of modeling and probing the complex dynamics of hemostatic responses in the native context of human blood vessels. To address this major challenge, this paper describes a microengineering approach for in vitro modeling of hemostasis. This microphysiological model replicates the living endothelium, multilayered microarchitecture, and procoagulant activity of human blood vessels, and is also equipped with a microneedle that is actuated with spatial precision to simulate penetrating vascular injuries. The system recapitulates key features of the hemostatic response to acute vascular injury as observed in vivo, including i) thrombin-driven accumulation of platelets and fibrin, ii) formation of a platelet- and fibrin-rich hemostatic plug that halts blood loss, and iii) matrix deformation driven by platelet contraction for wound closure. Moreover, the potential use of this model for drug testing applications is demonstrated by evaluating the effects of anticoagulants and antiplatelet agents that are in current clinical use. The vascular injury-on-a-chip may serve as an enabling platform for preclinical investigation of hematological disorders and emerging therapeutic approaches against them.
Collapse
Affiliation(s)
| | - Keon Woo Kwon
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jeongyun Seo
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Maurizio Tomaiuolo
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Timothy J Stalker
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lawrence F Brass
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute of Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
10
|
Barrachina MN, Izquierdo I, Hermida-Nogueira L, Morán LA, Pérez A, Arroyo AB, García-Barberá N, González-Conejero R, Troitiño S, Eble JA, Rivera J, Martínez C, Loza MI, Domínguez E, García Á. The PI3Kδ Inhibitor Idelalisib Diminishes Platelet Function and Shows Antithrombotic Potential. Int J Mol Sci 2021; 22:ijms22073304. [PMID: 33804911 PMCID: PMC8037016 DOI: 10.3390/ijms22073304] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/16/2021] [Accepted: 03/19/2021] [Indexed: 12/27/2022] Open
Abstract
Background: Clinical management of ischemic events and prevention of vascular disease is based on antiplatelet drugs. Given the relevance of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) as a candidate target in thrombosis, the main goal of the present study was to identify novel antiplatelet agents within the existing inhibitors blocking PI3K isoforms. Methods: We performed a biological evaluation of the pharmacological activity of PI3K inhibitors in platelets. The effect of the inhibitors was evaluated in intracellular calcium release and platelet functional assays, the latter including aggregation, adhesion, and viability assays. The in vivo drug antithrombotic potential was assessed in mice undergoing chemically induced arterial occlusion, and the associated hemorrhagic risk evaluated by measuring the tail bleeding time. Results: We show that PI3K Class IA inhibitors potently block calcium mobilization in human platelets. The PI3K p110δ inhibitor Idelalisib inhibits platelet aggregation mediated by ITAM receptors GPVI and CLEC-2, preferentially by the former. Moreover, Idelalisib also inhibits platelet adhesion and aggregation under shear and adhesion to collagen. Interestingly, an antithrombotic effect was observed in mice treated with Idelalisib, with mild bleeding effects at high doses of the drug. Conclusion: Idelalisib may have antiplatelet effects with minor bleeding effects, which provides a rationale to evaluate its antithrombotic efficacy in humans.
Collapse
Affiliation(s)
- María N. Barrachina
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases, Universidade Santiago de Compostela and Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain; (M.N.B.); (I.I.); (L.H.-N.); (L.A.M.); (S.T.)
| | - Irene Izquierdo
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases, Universidade Santiago de Compostela and Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain; (M.N.B.); (I.I.); (L.H.-N.); (L.A.M.); (S.T.)
| | - Lidia Hermida-Nogueira
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases, Universidade Santiago de Compostela and Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain; (M.N.B.); (I.I.); (L.H.-N.); (L.A.M.); (S.T.)
| | - Luis A. Morán
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases, Universidade Santiago de Compostela and Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain; (M.N.B.); (I.I.); (L.H.-N.); (L.A.M.); (S.T.)
| | - Amparo Pérez
- Pharmacology Applied to Drug Discovery Group, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas, Universidade Santiago de Compostela, 15705 Santiago de Compostela, Spain; (A.P.); (M.I.L.); (E.D.)
- Grupo Biofarma, Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain
| | - Ana B. Arroyo
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, 30003 Murcia, Spain; (A.B.A.); (N.G.-B.); (R.G.-C.); (J.R.); (C.M.)
| | - Nuria García-Barberá
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, 30003 Murcia, Spain; (A.B.A.); (N.G.-B.); (R.G.-C.); (J.R.); (C.M.)
| | - Rocío González-Conejero
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, 30003 Murcia, Spain; (A.B.A.); (N.G.-B.); (R.G.-C.); (J.R.); (C.M.)
| | - Sara Troitiño
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases, Universidade Santiago de Compostela and Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain; (M.N.B.); (I.I.); (L.H.-N.); (L.A.M.); (S.T.)
| | - Johannes A. Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany;
| | - José Rivera
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, 30003 Murcia, Spain; (A.B.A.); (N.G.-B.); (R.G.-C.); (J.R.); (C.M.)
| | - Constantino Martínez
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, 30003 Murcia, Spain; (A.B.A.); (N.G.-B.); (R.G.-C.); (J.R.); (C.M.)
| | - María I. Loza
- Pharmacology Applied to Drug Discovery Group, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas, Universidade Santiago de Compostela, 15705 Santiago de Compostela, Spain; (A.P.); (M.I.L.); (E.D.)
- Grupo Biofarma, Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain
| | - Eduardo Domínguez
- Pharmacology Applied to Drug Discovery Group, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas, Universidade Santiago de Compostela, 15705 Santiago de Compostela, Spain; (A.P.); (M.I.L.); (E.D.)
- Grupo Biofarma, Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain
| | - Ángel García
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases, Universidade Santiago de Compostela and Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain; (M.N.B.); (I.I.); (L.H.-N.); (L.A.M.); (S.T.)
- Correspondence: ; Tel.: +34-881-815429
| |
Collapse
|
11
|
Bray MA, Sartain SE, Gollamudi J, Rumbaut RE. Microvascular thrombosis: experimental and clinical implications. Transl Res 2020; 225:105-130. [PMID: 32454092 PMCID: PMC7245314 DOI: 10.1016/j.trsl.2020.05.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/12/2020] [Accepted: 05/17/2020] [Indexed: 02/07/2023]
Abstract
A significant amount of clinical and research interest in thrombosis is focused on large vessels (eg, stroke, myocardial infarction, deep venous thrombosis, etc.); however, thrombosis is often present in the microcirculation in a variety of significant human diseases, such as disseminated intravascular coagulation, thrombotic microangiopathy, sickle cell disease, and others. Further, microvascular thrombosis has recently been demonstrated in patients with COVID-19, and has been proposed to mediate the pathogenesis of organ injury in this disease. In many of these conditions, microvascular thrombosis is accompanied by inflammation, an association referred to as thromboinflammation. In this review, we discuss endogenous regulatory mechanisms that prevent thrombosis in the microcirculation, experimental approaches to induce microvascular thrombi, and clinical conditions associated with microvascular thrombosis. A greater understanding of the links between inflammation and thrombosis in the microcirculation is anticipated to provide optimal therapeutic targets for patients with diseases accompanied by microvascular thrombosis.
Collapse
Key Words
- adamts13, a disintegrin-like and metalloproteinase with thrombospondin type 1 motif 13
- ap, alternate pathway
- apc, activated protein c
- aps, antiphospholipid syndrome
- caps, catastrophic aps
- asfa, american society for apheresis
- atp, adenosine triphosphate
- cfh, complement factor h
- con a, concavalin a
- cox, cyclooxygenase
- damp, damage-associated molecular pattern
- dic, disseminated intravascular coagulation
- gbm, glomerular basement membrane
- hellp, hemolysis, elevated liver enzymes, low platelets
- hitt, heparin-induced thrombocytopenia and thrombosis
- hlh, hemophagocytic lymphohistiocytosis
- hus, hemolytic-uremic syndrome
- isth, international society for thrombosis and haemostasis
- ivig, intravenous immunoglobulin
- ldh, lactate nos, nitric oxide synthase
- net, neutrophil extracellular trap
- pai-1, plasminogen activator inhibitor 1
- pf4, platelet factor 4
- prr, pattern recognition receptor
- rbc, red blood cell
- scd, sickle cell disease
- sle, systemic lupus erythematosus
- tlr, toll-like receptor
- tf, tissue factor
- tfpi, tissue factor pathway inhibitor
- tma, thrombotic microangiopathy
- tnf-α, tumor necrosis factor-α
- tpe, therapeutic plasma exchange
- ulc, ultra large heparin-pf4 complexes
- ulvwf, ultra-large von willebrand factor
- vwf, von willebrand factor
Collapse
Affiliation(s)
- Monica A Bray
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, Texas; Baylor College of Medicine, Houston, Texas
| | - Sarah E Sartain
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, Texas; Baylor College of Medicine, Houston, Texas
| | - Jahnavi Gollamudi
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, Texas; Baylor College of Medicine, Houston, Texas
| | - Rolando E Rumbaut
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey VA Medical Center, Houston, Texas; Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
12
|
Donat C, Kölm R, Csorba K, Tuncer E, Tsakiris DA, Trendelenburg M. Complement C1q Enhances Primary Hemostasis. Front Immunol 2020; 11:1522. [PMID: 32765527 PMCID: PMC7381122 DOI: 10.3389/fimmu.2020.01522] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
The cross-talk between the inflammatory complement system and hemostasis is becoming increasingly recognized. The interaction between complement C1q, initiation molecule of the classical pathway, and von Willebrand factor (vWF), initiator molecule of primary hemostasis, has been shown to induce platelet rolling and adhesion in vitro. As vWF disorders result in prolonged bleeding, a lack of C1q as binding partner for vWF might also lead to an impaired hemostasis. Therefore, this study aimed to investigate the in vivo relevance of C1q-dependent binding of vWF in hemostasis. For this purpose, we analyzed parameters of primary and secondary hemostasis and performed bleeding experiments in wild type (WT) and C1q-deficient (C1qa−/−) mice, with reconstitution experiments of C1q in the latter. Bleeding tendency was examined by quantification of bleeding time and blood loss. First, we found that complete blood counts and plasma vWF levels do not differ between C1qa−/− mice and WT mice. Moreover, platelet aggregation tests indicated that the platelets of both strains of mice are functional. Second, while the prothrombin time was comparable between both groups, the activated partial thromboplastin time was shorter in C1qa−/− mice. In contrast, tail bleeding times of C1qa−/− mice were prolonged accompanied by an increased blood loss. Upon reconstitution of C1qa−/− mice with C1q, parameters of increased bleeding could be reversed. In conclusion, our data indicate that C1q, a molecule of the first-line of immune defense, actively participates in primary hemostasis by promoting arrest of bleeding. This observation might be of relevance for the understanding of thromboembolic complications in inflammatory disorders, where excess of C1q deposition is observed.
Collapse
Affiliation(s)
- Claudia Donat
- Laboratory of Clinical Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Robert Kölm
- Laboratory of Clinical Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Kinga Csorba
- Laboratory of Clinical Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Eylul Tuncer
- Laboratory of Clinical Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Dimitrios A Tsakiris
- Department of Diagnostic Hematology, University Hospital Basel, Basel, Switzerland
| | - Marten Trendelenburg
- Laboratory of Clinical Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland.,Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
13
|
Paul DS, Bergmeier W. Novel Mouse Model for Studying Hemostatic Function of Human Platelets. Arterioscler Thromb Vasc Biol 2020; 40:1891-1904. [PMID: 32493172 DOI: 10.1161/atvbaha.120.314304] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Platelets are critical to the formation of a hemostatic plug and the pathogenesis of atherothrombosis. Preclinical animal models, especially the mouse, provide an important platform to assess the efficacy and safety of antiplatelet drugs. However, these studies are limited by inherent differences between human and mouse platelets and the species-selectivity of many drugs. To circumvent these limitations, we developed a new protocol for the adoptive transfer of human platelets into thrombocytopenic nonobese diabetic/severe combined immune deficiency mice, that is, a model where all endogenous platelets are replaced by human platelets in mice accepting xenogeneic tissues. Approach and Results: To demonstrate the power of this new model, we visualized and quantified hemostatic plug formation and stability by intravital spinning disk confocal microscopy following laser ablation injury to the saphenous vein. Integrin αIIbβ3-dependent hemostatic platelet plug formation was achieved within ≈30 seconds after laser ablation injury in humanized platelet mice. Pretreatment of mice with standard dual antiplatelet therapy (Aspirin+Ticagrelor) or PAR1 inhibitor, L-003959712 (an analog of vorapaxar), mildly prolonged the bleeding time and significantly reduced platelet adhesion to the site of injury. Consistent with findings from clinical trials, inhibition of PAR1 in combination with dual antiplatelet therapy markedly prolonged bleeding time in humanized platelet mice. CONCLUSIONS We propose that this novel mouse model will provide a robust platform to test and predict the safety and efficacy of experimental antiplatelet drugs and to characterize the hemostatic function of synthetic, stored and patient platelets.
Collapse
Affiliation(s)
- David S Paul
- From the Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill (D.S.P., W.B.).,UNC Blood Research Center, University of North Carolina, Chapel Hill (D.S.P., W.B.)
| | - Wolfgang Bergmeier
- From the Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill (D.S.P., W.B.).,UNC Blood Research Center, University of North Carolina, Chapel Hill (D.S.P., W.B.)
| |
Collapse
|
14
|
Eriksson O, Mohlin C, Nilsson B, Ekdahl KN. The Human Platelet as an Innate Immune Cell: Interactions Between Activated Platelets and the Complement System. Front Immunol 2019; 10:1590. [PMID: 31354729 PMCID: PMC6635567 DOI: 10.3389/fimmu.2019.01590] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022] Open
Abstract
Platelets play an essential role in maintaining homeostasis in the circulatory system after an injury by forming a platelet thrombus, but they also occupy a central node in the intravascular innate immune system. This concept is supported by their extensive interactions with immune cells and the cascade systems of the blood. In this review we discuss the close relationship between platelets and the complement system and the role of these interactions during thromboinflammation. Platelets are protected from complement-mediated damage by soluble and membrane-expressed complement regulators, but they bind several complement components on their surfaces and trigger complement activation in the fluid phase. Furthermore, localized complement activation may enhance the procoagulant responses of platelets through the generation of procoagulant microparticles by insertion of sublytic amounts of C5b9 into the platelet membrane. We also highlight the role of post-translational protein modifications in regulating the complement system and the critical role of platelets in driving these reactions. In particular, modification of disulfide bonds by thiol isomerases and protein phosphorylation by extracellular kinases have emerged as important mechanisms to fine-tune complement activity in the platelet microenvironment. Lastly, we describe disorders with perturbed complement activation where part of the clinical presentation includes uncontrolled platelet activation that results in thrombocytopenia, and illustrate how complement-targeting drugs are alleviating the prothrombotic phenotype in these patients. Based on these clinical observations, we discuss the role of limited complement activation in enhancing platelet activation and consider how these drugs may provide opportunities for further dissecting the complex interactions between complement and platelets.
Collapse
Affiliation(s)
- Oskar Eriksson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Camilla Mohlin
- Linnaeus Center of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Kristina N. Ekdahl
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Linnaeus Center of Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| |
Collapse
|
15
|
Laurent PA, Hechler B, Solinhac R, Ragab A, Cabou C, Anquetil T, Severin S, Denis CV, Mangin PH, Vanhaesebroeck B, Payrastre B, Gratacap MP. Impact of PI3Kα (Phosphoinositide 3-Kinase Alpha) Inhibition on Hemostasis and Thrombosis. Arterioscler Thromb Vasc Biol 2018; 38:2041-2053. [PMID: 30354258 DOI: 10.1161/atvbaha.118.311410] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective- PI3Kα (phosphoinositide 3-kinase alpha) is a therapeutic target in oncology, but its role in platelets and thrombosis remains ill characterized. In this study, we have analyzed the role of PI3Kα in vitro, ex vivo, and in vivo in 2 models of arterial thrombosis. Approach and Results- Using mice selectively deficient in p110α in the megakaryocyte lineage and isoform-selective inhibitors, we confirm that PI3Kα is not mandatory but participates to thrombus growth over a collagen matrix at arterial shear rate. Our data uncover a role for PI3Kα in low-level activation of the GP (glycoprotein) VI-collagen receptor by contributing to ADP secretion and in turn full activation of PI3Kβ and Akt/PKB (protein kinase B). This effect was no longer observed at high level of GP VI agonist concentration. Our study also reveals that over a vWF (von Willebrand factor) matrix, PI3Kα regulates platelet stationary adhesion contacts under arterial flow through its involvement in the outside-in signaling of vWF-engaged αIIbβ3 integrin. In vivo, absence or inhibition of PI3Kα resulted in a modest but significant decrease in thrombus size after superficial injuries of mouse mesenteric arteries and an increased time to arterial occlusion after carotid lesion, without modification in the tail bleeding time. Considering the more discrete and nonredundant role of PI3Kα compared with PI3Kβ, selective PI3Kα inhibitors are unlikely to increase the bleeding risk at least in the absence of combination with antiplatelet drugs or thrombopenia. Conclusions- This study provides mechanistic insight into the role of PI3Kα in platelet activation and arterial thrombosis.
Collapse
Affiliation(s)
- Pierre-Alexandre Laurent
- From the INSERM, UMR-S1048, Université Toulouse III, France (P.-A.L., R.S., A.R., C.C., T.A., S.S., B.P., M.-P.G.)
| | - Béatrice Hechler
- INSERM, EFS Grand Est, BPPS UMR-S 949, FMTS, Université de Strasbourg, France (B.H., P.H.M.)
| | - Romain Solinhac
- From the INSERM, UMR-S1048, Université Toulouse III, France (P.-A.L., R.S., A.R., C.C., T.A., S.S., B.P., M.-P.G.)
| | - Ashraf Ragab
- From the INSERM, UMR-S1048, Université Toulouse III, France (P.-A.L., R.S., A.R., C.C., T.A., S.S., B.P., M.-P.G.)
| | - Cendrine Cabou
- From the INSERM, UMR-S1048, Université Toulouse III, France (P.-A.L., R.S., A.R., C.C., T.A., S.S., B.P., M.-P.G.)
| | - Typhaine Anquetil
- From the INSERM, UMR-S1048, Université Toulouse III, France (P.-A.L., R.S., A.R., C.C., T.A., S.S., B.P., M.-P.G.)
| | - Sonia Severin
- From the INSERM, UMR-S1048, Université Toulouse III, France (P.-A.L., R.S., A.R., C.C., T.A., S.S., B.P., M.-P.G.)
| | - Cécile V Denis
- INSERM, UMR-S 1176, University of Paris-Sud, Université Paris-Saclay, France (C.V.D.)
| | - Pierre H Mangin
- INSERM, EFS Grand Est, BPPS UMR-S 949, FMTS, Université de Strasbourg, France (B.H., P.H.M.)
| | - Bart Vanhaesebroeck
- Cell Signaling, UCL Cancer Institute, University College London, United Kingdom (B.V.)
| | - Bernard Payrastre
- From the INSERM, UMR-S1048, Université Toulouse III, France (P.-A.L., R.S., A.R., C.C., T.A., S.S., B.P., M.-P.G.)
- CHU de Toulouse, Laboratoire d'Hématologie, France (B.P.)
| | - Marie-Pierre Gratacap
- From the INSERM, UMR-S1048, Université Toulouse III, France (P.-A.L., R.S., A.R., C.C., T.A., S.S., B.P., M.-P.G.)
| |
Collapse
|
16
|
Iegre J, Ahmed NS, Gaynord JS, Wu Y, Herlihy KM, Tan YS, Lopes-Pires ME, Jha R, Lau YH, Sore HF, Verma C, O' Donovan DH, Pugh N, Spring DR. Stapled peptides as a new technology to investigate protein-protein interactions in human platelets. Chem Sci 2018; 9:4638-4643. [PMID: 29899957 PMCID: PMC5969508 DOI: 10.1039/c8sc00284c] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/23/2018] [Indexed: 12/14/2022] Open
Abstract
We describe the first application of stapled peptides in human platelets. Bim BH3 stapled peptides are used to overcome the limitations of traditional methods and uncover a new role for Bim in platelet activation.
Platelets are blood cells with numerous crucial pathophysiological roles in hemostasis, cardiovascular thrombotic events and cancer metastasis. Platelet activation requires the engagement of intracellular signalling pathways that involve protein–protein interactions (PPIs). A better understanding of these pathways is therefore crucial for the development of selective anti-platelet drugs. New strategies for studying PPIs in human platelets are required to overcome limitations associated with conventional platelet research methods. For example, small molecule inhibitors can lack selectivity and are often difficult to design and synthesise. Additionally, development of transgenic animal models is costly and time-consuming and conventional recombinant techniques are ineffective due to the lack of a nucleus in platelets. Herein, we describe the generation of a library of novel, functionalised stapled peptides and their first application in the investigation of platelet PPIs. Moreover, the use of platelet-permeable stapled Bim BH3 peptides confirms the part of Bim in phosphatidyl-serine (PS) exposure and reveals a role for the Bim protein in platelet activatory processes. Our work demonstrates that functionalised stapled peptides are a complementary alternative to conventional platelet research methods, and could make a significant contribution to the understanding of platelet signalling pathways and hence to the development of anti-platelet drugs.
Collapse
Affiliation(s)
- Jessica Iegre
- Department of Chemistry , University of Cambridge , CB2 1EW , UK .
| | - Niaz S Ahmed
- Department of Biomedical and Forensic Sciences , Anglia Ruskin University , CB1 1PT , UK .
| | | | - Yuteng Wu
- Department of Chemistry , University of Cambridge , CB2 1EW , UK .
| | - Kara M Herlihy
- Discovery Sciences , IMED Biotech Unit , AstraZeneca , Cambridge , UK
| | - Yaw Sing Tan
- Bioinformatics Institute , Agency for Science, Technology and Research (ASTAR) , 30 Biopolis Street, #07-01 Matrix , 13867 , Singapore
| | - Maria E Lopes-Pires
- Department of Biomedical and Forensic Sciences , Anglia Ruskin University , CB1 1PT , UK .
| | - Rupam Jha
- Discovery Sciences , IMED Biotech Unit , AstraZeneca , Cambridge , UK
| | - Yu Heng Lau
- Department of Chemistry , University of Cambridge , CB2 1EW , UK . .,School of Chemistry , The University of Sydney , NSW 2006 , Australia
| | - Hannah F Sore
- Department of Chemistry , University of Cambridge , CB2 1EW , UK .
| | - Chandra Verma
- Bioinformatics Institute , Agency for Science, Technology and Research (ASTAR) , 30 Biopolis Street, #07-01 Matrix , 13867 , Singapore.,Department of Biological Sciences , National University of Singapore , 14 Science Drive 4 , Singapore 117543.,School of Biological Sciences , Nanyang Technological University , 60 Nanyang Drive , Singapore 637551
| | | | - Nicholas Pugh
- Department of Biomedical and Forensic Sciences , Anglia Ruskin University , CB1 1PT , UK .
| | - David R Spring
- Department of Chemistry , University of Cambridge , CB2 1EW , UK .
| |
Collapse
|
17
|
Sánchez G, Estrada O, Acha G, Cardozo A, Peña F, Ruiz MC, Michelangeli F, Alvarado-Castillo C. The norpurpureine alkaloid from Annona purpurea inhibits human platelet activation in vitro. Cell Mol Biol Lett 2018; 23:15. [PMID: 29713353 PMCID: PMC5905151 DOI: 10.1186/s11658-018-0082-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 04/09/2018] [Indexed: 01/01/2023] Open
Abstract
Background The leaves of Annona purpurea have yielded several alkaloids with anti-aggregation activities against rabbit platelets. This is promising in the search for agents that might act against platelets and reduce the incidence of cardiovascular diseases. Since significant differences in platelet function have been reported between human and animal platelets, a study focusing on the effect of A. purpurea extracts against human platelet activation is necessary. Methods The compounds in an A. purpurea ethanolic extract underwent bio-guided fractionation and were used for in vitro human platelet aggregation assays to isolate the compounds with anti-platelet activity. The bioactive compounds were identified by spectroscopic analysis. Additional platelet studies were performed to characterize their action as inhibitors of human platelet activation. Results The benzylisoquinoline alkaloid norpurpureine was identified as the major anti-platelet compound. The IC50 for norpurpureine was 80 μM against platelets when stimulated with adenosine 5′-diphosphate (ADP), collagen and thrombin. It was pharmacologically effective from 20 to 220 μM. Norpurpureine (220 μM) exhibited its in vitro effectiveness in samples from 30 healthy human donors who did not take any drugs during the 2 weeks prior to the collection. Norpurpureine also gradually inhibited granule secretion and adhesion of activated platelets to immobilized fibrinogen. At the intra-platelet level, norpurpureine prevented agonist-stimulated calcium mobilization and cAMP reduction. Structure–activity relationship analysis indicates that the lack of a methyl group at the nitrogen seems to be key in the ability of the compound to interact with its molecular target. Conclusion Norpurpureine displays a promising in vitro pharmacological profile as an inhibitor of human platelet activation. Its molecular target could be a common effector between Ca2+ and cAMP signaling, such as the PLC-PKC-Ca2+ pathway and PDEs. This needs further evaluation at the protein isoform level. Electronic supplementary material The online version of this article (10.1186/s11658-018-0082-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gabriela Sánchez
- 1Centro de Biofísica y Bioquímica (CBB), Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Bolivarian Republic of Venezuela
| | - Omar Estrada
- 1Centro de Biofísica y Bioquímica (CBB), Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Bolivarian Republic of Venezuela
| | - Giovana Acha
- 1Centro de Biofísica y Bioquímica (CBB), Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Bolivarian Republic of Venezuela
| | - Alfonso Cardozo
- 2Laboratorio de Botánica Sistemática, Facultad de Agronomía, Universidad Central de Venezuela (UCV), Maracay, Bolivarian Republic of Venezuela
| | - Franshelle Peña
- 1Centro de Biofísica y Bioquímica (CBB), Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Bolivarian Republic of Venezuela
| | - Marie Christine Ruiz
- 1Centro de Biofísica y Bioquímica (CBB), Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Bolivarian Republic of Venezuela
| | - Fabián Michelangeli
- 1Centro de Biofísica y Bioquímica (CBB), Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Bolivarian Republic of Venezuela
| | - Claudia Alvarado-Castillo
- 1Centro de Biofísica y Bioquímica (CBB), Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Bolivarian Republic of Venezuela.,3Laboratorio de Hemostasia y Genética Vascular, Centro de Biofísica y Bioquímica, Instituto Venezolano de Investigaciones Científicas, Apartado 20632, K11 de la Carretera Panamericana, Caracas, 1020-A Bolivarian Republic of Venezuela
| |
Collapse
|
18
|
Novotny J, Chandraratne S, Weinberger T, Philippi V, Stark K, Ehrlich A, Pircher J, Konrad I, Oberdieck P, Titova A, Hoti Q, Schubert I, Legate KR, Urtz N, Lorenz M, Pelisek J, Massberg S, von Brühl ML, Schulz C. Histological comparison of arterial thrombi in mice and men and the influence of Cl-amidine on thrombus formation. PLoS One 2018; 13:e0190728. [PMID: 29293656 PMCID: PMC5749862 DOI: 10.1371/journal.pone.0190728] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 12/19/2017] [Indexed: 02/08/2023] Open
Abstract
Aims Medical treatment of arterial thrombosis is mainly directed against platelets and coagulation factors, and can lead to bleeding complications. Novel antithrombotic therapies targeting immune cells and neutrophil extracellular traps (NETs) are currently being investigated in animals. We addressed whether immune cell composition of arterial thrombi induced in mouse models of thrombosis resemble those of human patients with acute myocardial infarction (AMI). Methods and results In a prospective cohort study of patients suffering from AMI, 81 human arterial thrombi were harvested during percutaneous coronary intervention and subjected to detailed histological analysis. In mice, arterial thrombi were induced using two distinct experimental models, ferric chloride (FeCl3) and wire injury of the carotid artery. We found that murine arterial thrombi induced by FeCl3 were highly concordant with human coronary thrombi regarding their immune cell composition, with neutrophils being the most abundant cell type, as well as the presence of NETs and coagulation factors. Pharmacological treatment of mice with the protein arginine deiminase (PAD)-inhibitor Cl-amidine abrogated NET formation, reduced arterial thrombosis and limited injury in a model of myocardial infarction. Conclusions Neutrophils are a hallmark of arterial thrombi in patients suffering from acute myocardial infarction and in mouse models of arterial thrombosis. Inhibition of PAD could represent an interesting strategy for the treatment of arterial thrombosis to reduce neutrophil-associated tissue damage and improve functional outcome.
Collapse
Affiliation(s)
- Julia Novotny
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Sue Chandraratne
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Tobias Weinberger
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Vanessa Philippi
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Konstantin Stark
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Andreas Ehrlich
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Joachim Pircher
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Ildiko Konrad
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Paul Oberdieck
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Anna Titova
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Qendresa Hoti
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Irene Schubert
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Kyle R. Legate
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Department of Applied Physics, Center for NanoSciences, Ludwig-Maximilians-Universität, Munich, Germany
| | - Nicole Urtz
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Michael Lorenz
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Jaroslav Pelisek
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar der Technischen Universität, Munich, Germany
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Marie-Luise von Brühl
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, Ludwig-Maximilians-Universität, Munich, Germany
- Walter-Brendel-Centre of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- * E-mail:
| |
Collapse
|
19
|
Enayat S, Ravanbod S, Rassoulzadegan M, Jazebi M, Tarighat S, Ala F, Emsley J, Othman M. A novel D235Y mutation in the GP1BA gene enhances platelet interaction with von Willebrand factor in an Iranian family with platelet-type von Willebrand disease. Thromb Haemost 2017; 108:946-54. [DOI: 10.1160/th12-04-0189] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 08/08/2012] [Indexed: 11/05/2022]
Abstract
SummaryPlatelet-type von Willebrand disease (PT-VWD) is a rare bleeding disorder with an intrinsic defect in platelets rather than von Willebrand factor (VWF), but has clinical and laboratory features similar to the more common type 2B VWD. The intriguing nature of the pathophysiology and molecular genetics of PT-VWD has created lengthy debate in literature regarding its discrimination from type 2B VWD, and essentially confirming DNA analysis as the gold standard in diagnosis and revealing pathologic mutations. In this report we identify a novel Asp235Tyr mutation in the GP1BA gene of two Iranian patients showing the PT-VWD phenotype who were originally misdiagnosed as type 2B VWD. By structural modelling of the mutant by introducing Tyr235 into the available crystal structure of the glycoprotein (GP)Ibα N-terminal domain, we observed the mutant Tyr235 generates a hydrophobic tip to the extended β-switch loop of GPIbα. Further modelling of the resulting complex with VWFA1 indicates this could result in an enhanced interface compared to wild-type Asp235. This data provides an update to the present knowledge about this rare disorder, and confirms the necessity of genetic testing for accurate diagnosis, and the importance of studying natural mutations to better understand molecular aspects of GPIbα-VWFA1 interaction.
Collapse
|
20
|
Fröhlich E. Action of Nanoparticles on Platelet Activation and Plasmatic Coagulation. Curr Med Chem 2016; 23:408-30. [PMID: 26063498 PMCID: PMC5403968 DOI: 10.2174/0929867323666160106151428] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 12/01/2015] [Accepted: 01/05/2016] [Indexed: 12/14/2022]
Abstract
Nanomaterials can get into the blood circulation after injection or by release from implants but also by permeation of the epithelium after oral, respiratory or dermal exposure. Once in the blood, they can affect hemostasis, which is usually not intended. This review addresses effects of biological particles and engineered nanomaterials on hemostasis. The role of platelets and coagulation in normal clotting and the interaction with the immune system are described. Methods to identify effects of nanomaterials on clotting and results from in vitro and in vivo studies are summarized and the role of particle size and surface properties discussed. The literature overview showed that mainly pro-coagulative effects of nanomaterials have been described. In vitro studies suggested stronger effects of smaller than of larger NPs on coagulation and a greater importance of material than of surface charge. For instance, carbon nanotubes, polystyrene particles, and dendrimers inferred with clotting independent from their surface charge. Coating of particles with polyethylene glycol was able to prevent interaction with clotting by some particles, while it had no effect on others and the more recently developed bio-inspired surfaces might help to design coatings for more biocompatible particles. The mainly pro-coagulative action of nanoparticles could present a particular risk for individuals affected by common diseases such as diabetes, cancer, and cardiovascular diseases. Under standardized conditions, in vitro assays using human blood appear to be a suitable tool to study mechanisms of interference with hemostasis and to optimize hemocompatibility of nanomaterials.
Collapse
Affiliation(s)
- Eleonore Fröhlich
- Center for Medical Research, Medical University Graz, Stiftingtalstr 24, 8010 Graz, Austria.
| |
Collapse
|
21
|
Kamat V, Muthard RW, Li R, Diamond SL. Microfluidic assessment of functional culture-derived platelets in human thrombi under flow. Exp Hematol 2015; 43:891-900.e4. [PMID: 26145051 DOI: 10.1016/j.exphem.2015.06.302] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 06/12/2015] [Accepted: 06/25/2015] [Indexed: 11/30/2022]
Abstract
Despite their clinical significance, human platelets are not amenable to genetic manipulation, thus forcing a reliance on mouse models. Culture-derived platelets (CDPs) from human peripheral blood CD34(+) cells can be genetically altered and may eventually be used for transfusions. By use of microfluidics, the time-dependent incorporation of CD41(+)CD42(+) CDPs into clots was measured using only 54,000 CDPs doped into 27 μL of human whole blood perfused over collagen at a wall shear rate of 100 sec(-1). With the use of fluorescence-labeled human platelets (instead of CDPs) doped between 0.25% and 2% of total platelets, incorporation was highly quantitative and allowed monitoring of the anti-αIIbβ3 antagonism that occurred after collagen adhesion. CDPs were only 15% as efficient as human platelets in their incorporation into human thrombi under flow, although both cell types were equally antagonized by αIIbβ3 inhibition. Transient transfection allowed the monitoring of GFP(+) human CDP incorporation into clots. This assay quantifies genetically altered CDP function under flow.
Collapse
Affiliation(s)
- Viraj Kamat
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ryan W Muthard
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ruizhi Li
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Scott L Diamond
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
22
|
Platelets at the interface of thrombosis, inflammation, and cancer. Blood 2015; 126:582-8. [PMID: 26109205 DOI: 10.1182/blood-2014-08-531582] [Citation(s) in RCA: 461] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 06/18/2015] [Indexed: 12/13/2022] Open
Abstract
Although once primarily recognized for its roles in hemostasis and thrombosis, the platelet has been increasingly recognized as a multipurpose cell. Indeed, circulating platelets have the ability to influence a wide range of seemingly unrelated pathophysiologic events. Here, we highlight some of the notable observations that link platelets to inflammation, reinforcing the platelet's origin from a lower vertebrate cell type with both hemostatic and immunologic roles. In addition, we consider the relevance of platelets in cancer biology by focusing on the hallmarks of cancer and the ways platelets can influence multistep development of tumors. Beyond its traditional role in hemostasis and thrombosis, the platelet's involvement in the interplay between hemostasis, thrombosis, inflammation, and cancer is likely complex, yet extremely important in each disease process. The existence of animal models of platelet dysfunction and currently used antiplatelet therapies provide a framework for understanding mechanistic insights into a wide range of pathophysiologic events. Thus, the basic scientist studying platelet function can think beyond the traditional hemostasis and thrombosis paradigms, while the practicing hematologist must appreciate platelet relevance in a wide range of disease processes.
Collapse
|
23
|
Ware J, Corken A. Platelets. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
24
|
LeVine DN, Birkenheuer AJ, Brooks MB, Nordone SK, Bellinger DA, Jones SL, Fischer TH, Oglesbee SE, Frey K, Brinson NS, Peters AP, Marr HS, Motsinger-Reif A, Gudbrandsdottir S, Bussel JB, Key NS. A novel canine model of immune thrombocytopenia: has immune thrombocytopenia (ITP) gone to the dogs? Br J Haematol 2014; 167:110-20. [PMID: 25039744 DOI: 10.1111/bjh.13005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 05/11/2014] [Indexed: 01/25/2023]
Abstract
Canine immune thrombocytopenia (ITP) is analogous to human ITP, with similar platelet counts and heterogeneity in bleeding phenotype among affected individuals. With a goal of ultimately investigating this bleeding heterogeneity, a canine model of antibody-mediated ITP was developed. Infusion of healthy dogs with 2F9, a murine IgG2a monoclonal antibody to the canine platelet glycoprotein GPIIb (a common target of autoantibodies in ITP) resulted in profound, dose-dependent thrombocytopenia. Model dogs developed variable bleeding phenotypes, e.g. petechiae and haematuria, despite similar degrees of thrombocytopenia. 2F9 infusion was not associated with systemic inflammation, consumptive coagulopathy, or impairment of platelet function. Unexpectedly however, evaluation of cytokine profiles led to the identification of platelets as a potential source of serum interleukin-8 (IL8) in dogs. This finding was confirmed in humans with ITP, suggesting that platelet IL8 may be a previously unrecognized modulator of platelet-neutrophil crosstalk. The utility of this model will allow future study of bleeding phenotypic heterogeneity including the role of neutrophils and endothelial cells in ITP.
Collapse
Affiliation(s)
- Dana N LeVine
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA; Department of Pathology and Laboratory Animal Medicine, University of North Carolina, Chapel Hill, NC, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
The role of microclot formation in an acute subarachnoid hemorrhage model in the rabbit. BIOMED RESEARCH INTERNATIONAL 2014; 2014:161702. [PMID: 25110658 PMCID: PMC4109416 DOI: 10.1155/2014/161702] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 06/02/2014] [Indexed: 12/21/2022]
Abstract
Background. Microvascular dysfunction and microthrombi formation are believed to contribute to development of early brain injury (EBI) after aneurysmal subarachnoid hemorrhage (SAH). Objective. This study aimed to determine (i) extent of microthrombus formation and neuronal apoptosis in the brain parenchyma using a blood shunt SAH model in rabbits; (ii) correlation of structural changes in microvessels with EBI characteristics. Methods. Acute SAH was induced using a rabbit shunt cisterna magna model. Extent of microthrombosis was detected 24 h post-SAH (n = 8) by fibrinogen immunostaining, compared to controls (n = 4). We assessed apoptosis by terminal deoxynucleotidyl transferase nick end labeling (TUNEL) in cortex and hippocampus. Results. Our results showed significantly more TUNEL-positive cells (SAH: 115 ± 13; controls: 58 ± 10; P = 0.016) and fibrinogen-positive microthromboemboli (SAH: 9 ± 2; controls: 2 ± 1; P = 0.03) in the hippocampus after aneurysmal SAH. Conclusions. We found clear evidence of early microclot formation in a rabbit model of acute SAH. The extent of microthrombosis did not correlate with early apoptosis or CPP depletion after SAH; however, the total number of TUNEL positive cells in the cortex and the hippocampus significantly correlated with mean CPP reduction during the phase of maximum depletion after SAH induction. Both microthrombosis and neuronal apoptosis may contribute to EBI and subsequent DCI.
Collapse
|
26
|
Chi X, Zhi L, Gelderman MP, Vostal JG. Host platelets and, in part, neutrophils mediate lung accumulation of transfused UVB-irradiated human platelets in a mouse model of acute lung injury. PLoS One 2012; 7:e44829. [PMID: 23028636 PMCID: PMC3446987 DOI: 10.1371/journal.pone.0044829] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 08/07/2012] [Indexed: 11/26/2022] Open
Abstract
We previously reported that ultraviolet light B (UVB)-treated human platelets (hPLTs) can cause acute lung injury (ALI) in a two-event SCID mouse model in which the predisposing event was Lipopolysaccharide (LPS) injection and the second event was infusion of UVB-treated hPLTs. To delineate contributions of host mouse platelets (mPLTs) and neutrophils in the pathogenesis of ALI in this mouse model, we depleted mPLTs or neutrophils and measured hPLT accumulation in the lung. We also assessed lung injury by protein content in bronchoalveolar lavage fluid (BALF). LPS injection followed by infusion of UVB-treated hPLTs resulted in sequestration of both mPLTs and hPLTs in the lungs of SCID mice, although the numbers of neutrophils in the lung were not significantly different from the control group. Depletion of mouse neutrophils caused only a mild reduction in UVB-hPLTs accumulation in the lungs and a mild reduction in protein content in BALF. In comparison, depletion of mPLTs almost completely abolished hPLTs accumulation in the lung and significantly reduced protein content in BALF. UVB-treated hPLTs bound to host mPLTs, but did not bind to neutrophils in the lung. Aspirin treatment of hPLTs in vitro abolished hPLT accumulation in the lung and protected mice from lung injury. Our data indicate that host mPLTs accumulated in the lungs in response to an inflammatory challenge and subsequently mediated the attachment of transfused UVB-hPLTs. Neutrophils also recruited a small percentage of platelets to the lung. These findings may help develop therapeutic strategies for ALI which could potentially result from transfusion of UV illuminated platelets.
Collapse
Affiliation(s)
| | | | | | - Jaroslav G. Vostal
- Laboratory of Cellular Hematology, Division of Hematology, OBRR, Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, Maryland, United States of America
- * E-mail:
| |
Collapse
|
27
|
Othman M, Lopez JA, Ware J. Platelet-type von Willebrand disease update: the disease, the molecule and the animal model. Expert Rev Hematol 2012; 4:475-7. [PMID: 21939413 DOI: 10.1586/ehm.11.45] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
28
|
Atherosclerosis and thrombosis: insights from large animal models. J Biomed Biotechnol 2011; 2011:907575. [PMID: 21274431 PMCID: PMC3022266 DOI: 10.1155/2011/907575] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 12/09/2010] [Indexed: 11/21/2022] Open
Abstract
Atherosclerosis and its thrombotic complications are responsible for remarkably high numbers of deaths. The combination of in vitro, ex vivo, and in vivo experimental approaches has largely contributed to a better understanding of the mechanisms underlying the atherothrombotic process. Indeed, different animal models have been implemented in atherosclerosis and thrombosis research in order to provide new insights into the mechanisms that have already been outlined in isolated cells and protein studies. Yet, although no model completely mimics the human pathology, large animal models have demonstrated better suitability for translation to humans. Indeed, direct translation from mice to humans should be taken with caution because of the well-reported species-related differences. This paper provides an overview of the available atherothrombotic-like animal models, with a particular focus on large animal models of thrombosis and atherosclerosis, and examines their applicability for translational research purposes as well as highlights species-related differences with humans.
Collapse
|
29
|
Guerrero JA, Kyei M, Russell S, Liu J, Gartner TK, Storrie B, Ware J. Visualizing the von Willebrand factor/glycoprotein Ib-IX axis with a platelet-type von Willebrand disease mutation. Blood 2009; 114:5541-6. [PMID: 19808696 PMCID: PMC2798865 DOI: 10.1182/blood-2009-03-210823] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 08/31/2009] [Indexed: 11/20/2022] Open
Abstract
Platelet-type von Willebrand disease (PT-VWD) is a bleeding disorder of the platelet glycoprotein Ib-IX/von Willebrand factor (VWF) axis caused by mutations in the glycoprotein Ib-IX receptor that lead to an increased affinity with VWF. In this report, platelets from a mouse expressing a mutation associated with PT-VWD have been visualized using state-of-the art image collection and processing. Confocal analysis revealed that VWF bound to the surface of single platelets and bridging micro-aggregates of platelets. Surface-bound VWF appears as a large, linear structure on the surface of 50% of the PT-VWD platelets. In vivo thrombus formation after chemical injury to the carotid artery revealed a severe impairment to occlusion as a consequence of the PT-VWD mutation. In vitro stimulation of PT-VWD platelets with adenosine diphosphate or thrombin demonstrates a significant block in their ability to bind fibrinogen. The impairment of in vivo thrombus formation and in vitro fibrinogen binding are more significant than might be expected from the observed platelet binding to VWF polymers over a small portion of the plasma membrane. Visualization of the receptor/ligand interaction and characterization of a severe antithrombotic phenotype provide a new understanding on the molecular basis of bleeding associated with the PT-VWD phenotype.
Collapse
Affiliation(s)
- Jose A Guerrero
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
During recent decades there have been major advances in the fields of thrombosis and haemostasis, in part through development of powerful molecular and genetic technologies. Nevertheless, genetic modification of megakaryocytes and generation of mutant platelets in vitro remains a highly specialized area of research. Developments are hampered by the low frequency of megakaryocytes and their progenitors, a poor efficiency of transfection and a lack of understanding with regard to the mechanism by which megakaryocytes release platelets. Current methods used in the generation of genetically modified megakaryocytes and platelets include mutant mouse models, cell line studies and use of viruses to transform primary megakaryocytes or haematopoietic precursor cells. This review summarizes the advantages, limitations and technical challenges of such methods, with a particular focus on recent successes and advances in this rapidly progressing field including the potential for use in gene therapy for treatment of patients with platelet disorders.
Collapse
Affiliation(s)
- Caroline Pendaries
- Centre for Cardiovascular Sciences, Institute for Biomedical Research, Wolfson Drive, The Medical School, University of Birmingham, Edgbaston, Birmingham, UK
| | | | | |
Collapse
|
31
|
Aljamali MN, Margaritis P, Schlachterman A, Tai SJ, Roy E, Bunte R, Camire RM, High KA. Long-term expression of murine activated factor VII is safe, but elevated levels cause premature mortality. J Clin Invest 2008; 118:1825-34. [PMID: 18398505 DOI: 10.1172/jci32878] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2007] [Accepted: 02/15/2008] [Indexed: 11/17/2022] Open
Abstract
Intravenous infusion of recombinant human activated Factor VII (FVIIa) has been used for over a decade in the successful management of bleeding episodes in patients with inhibitory antibodies to Factor VIII or Factor IX. Previously, we showed that expression of murine FVIIa (mFVIIa) from an adeno-associated viral (AAV) vector corrected abnormal hemostatic parameters in hemophilia B mice. To pursue this as a therapeutic approach, we sought to define safe and effective levels of FVIIa for continuous expression. In mice transgenic for mFVIIa or injected with AAV-mFVIIa, we analyzed survival, expression levels, in vitro and in vivo coagulation tests, and histopathology for up to 16 months after birth/mFVIIa expression. We found that continuous expression of mFVIIa at levels at or below 1.5 microg/ml was safe, effective, and compatible with a normal lifespan. However, expression levels of 2 microg/ml or higher were associated with thrombosis and early mortality, with pathologic findings in the heart and lungs that were rescued in a low-factor X (low-FX) mouse background, suggesting a FX-mediated effect. The findings from these mouse models of continuous FVIIa expression have implications for the development of a safe gene transfer approach for hemophilia and are consistent with the possibility of thromboembolic risk of continuously elevated FVIIa levels.
Collapse
Affiliation(s)
- Majed N Aljamali
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Suva LJ, Hartman E, Dilley JD, Russell S, Akel NS, Skinner RA, Hogue WR, Budde U, Varughese KI, Kanaji T, Ware J. Platelet dysfunction and a high bone mass phenotype in a murine model of platelet-type von Willebrand disease. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:430-9. [PMID: 18187573 DOI: 10.2353/ajpath.2008.070417] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The platelet glycoprotein Ib-IX receptor binds surface-bound von Willebrand factor and supports platelet adhesion to damaged vascular surfaces. A limited number of mutations within the glycoprotein Ib-IX complex have been described that permit a structurally altered receptor to interact with soluble von Willebrand factor, and this is the molecular basis of platelet-type von Willebrand disease. We have developed and characterized a mouse model of platelet-type von Willebrand disease (G233V) and have confirmed a platelet phenotype mimicking the human disorder. The mice have a dramatic increase in splenic megakaryocytes and splenomegaly. Recent studies have demonstrated that hematopoetic cells can influence the differentiation of osteogenic cells. Thus, we examined the skeletal phenotype of mice expressing the G233V variant complex. At 6 months of age, G233V mice exhibit a high bone mass phenotype with an approximate doubling of trabecular bone volume in both the tibia and femur. Serum measures of bone resorption were significantly decreased in G233V animals. With decreased bone resorption, cortical thickness was increased, medullary area decreased, and consequently, the mechanical strength of the femur was significantly increased. Using ex vivo bone marrow cultures, osteoclast-specific staining in the G233V mutant marrow was diminished, whereas osteoblastogenesis was unaffected. These studies provide new insights into the relationship between the regulation of megakaryocytopoiesis and bone mass.
Collapse
Affiliation(s)
- Larry J Suva
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Platelets play a central role in hemostasis, but also in atherothrombosis, as they rapidly adhere to tissue and to one another as a response to any vascular injury. This process involves a large number of surface receptors, signaling pathways, and enzymatic cascades as well as their complex interplay. Although in vitro experiments proved successful in both identifying new receptors and pathways and developing potent and selective antithrombotic drugs, in vitro research cannot mimic the myriad hemodynamic and spatiotemporal cellular and molecular interactions that occur during the generation and propagation of thrombi in vivo. Animal models, and, with the availability of genetically modified mouse strains and of modern intravital imaging techniques, mouse models in particular, have opened new ways to identify both individual roles and the interplay of platelet proteins in complex in vivo settings. In vivo models revealed the important role of, eg, Gas6 or blood coagulation factor XII in thrombus formation, and results obtained in in vivo models raised the interesting possibility that (physiologic) hemostasis and (pathologic) thrombosis might represent 2 mechanistically different processes. This review summarizes in vivo findings that contributed significantly to our understanding of hemostatic and thrombotic processes and which may help to guide future research.
Collapse
Affiliation(s)
- Ulrich J H Sachs
- Rudolf Virchow Center, Deutsche Forschungsgemeinschaft Research Center for Experimental Biomedicine, University of Würzburg, Germany
| | | |
Collapse
|
34
|
Denis CV, Wagner DD. Platelet adhesion receptors and their ligands in mouse models of thrombosis. Arterioscler Thromb Vasc Biol 2007; 27:728-39. [PMID: 17272754 DOI: 10.1161/01.atv.0000259359.52265.62] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Platelet adhesion and aggregation at sites of vascular injury are two key events in hemostasis and thrombosis. Because of exciting advances in genetic engineering, the mouse has become an important and frequently used model to unravel the molecular mechanisms underlying the multistep process leading to the formation of a stable platelet plug. In gene-targeted mice, the crucial importance of platelet adhesion receptors such as glycoprotein Ib alpha or the alphaIIb beta3 integrin has been confirmed and further clarified. Their absence leads to highly impaired thrombus formation, independent of the model used to induce vascular injury. In contrast, the relative contribution of other receptors, such as glycoprotein VI, or of various platelet ligands may be regulated by the severity of injury, the type of vessel injured, and the signaling pathways that are generated. Murine models have also helped improve understanding of the second wave of events that leads to stabilization of the platelet aggregate. Despite the current limitations due to lack of standardization and the virtual absence of thrombosis models in diseased vessels, there is no doubt that the mouse will play a key role in the discovery and characterization of the next generation of antithrombotic agents. This review focuses on key findings about the molecular mechanisms supporting hemostasis and thrombosis that have been obtained with genetically engineered mouse models deficient in various platelet adhesion receptors and ligands. Combination of these models with sophisticated methods allowing direct visualization of platelet-vessel wall interactions after injury greatly contributed to recent advances in the field.
Collapse
|
35
|
Wilcox DA, White GC. Gene Therapy for Platelet Disorders. Platelets 2007. [DOI: 10.1016/b978-012369367-9/50833-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
36
|
David T, Ohlmann P, Eckly A, Moog S, Cazenave JP, Gachet C, Lanza F. Inhibition of adhesive and signaling functions of the platelet GPIb-V-IX complex by a cell penetrating GPIbalpha peptide. J Thromb Haemost 2006; 4:2645-55. [PMID: 17100656 DOI: 10.1111/j.1538-7836.2006.02198.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Interaction between the platelet glycoprotein (GP)Ib-V-IX complex and von Willebrand factor (VWF) is critical for initiating platelet-vessel wall contacts, particularly under high shear conditions. This interaction also plays an important role in initiating platelet activation through the generation of intracellular signals resulting in platelet shape change and integrin alpha(IIb)beta3 activation. OBJECTIVE A cell-penetrating peptide strategy was used to study the role of the intracellular domain of the GPIbalpha subunit in VWF/GPIb-V-IX-dependent adhesion and activation. METHODS Peptides of 11-13 amino acids, covering the 557-610 region, were coupled to a nine-arginine permeating tag (R9) and the effects of their cell entry on VWF-dependent responses were analyzed. RESULTS The R9alpha557 peptide corresponding to the 557-569 segment reduced platelet agglutination in response to VWF, while the other peptides had no effect. The decreased platelet agglutination appeared to be an indirect consequence of adenosine diphosphate release as a normal response was restored by apyrase or a P2Y1 receptor antagonist. A more direct effect of R9alpha557 on GPIb VWF-dependent functions was observed in adhesion studies on a VWF matrix, where it decreased platelet adhesion and profoundly inhibited filopodia formation. In addition, cell adhesion was reduced and shape change absent when Chinese hamster ovary cells expressing the GPIb-IX complex were incubated with R9alpha557. CONCLUSION This study performed in intact platelets suggests a functional role of the 557-569 domain of GPIbalpha in controlling VWF-dependent adhesion and signaling.
Collapse
Affiliation(s)
- T David
- INSERM U311, Strasbourg; EFS-Alsace, Strasbourg, France
| | | | | | | | | | | | | |
Collapse
|
37
|
Strassel C, David T, Eckly A, Baas MJ, Moog S, Ravanat C, Trzeciak MC, Vinciguerra C, Cazenave JP, Gachet C, Lanza F. Synthesis of GPIb beta with novel transmembrane and cytoplasmic sequences in a Bernard-Soulier patient resulting in GPIb-defective signaling in CHO cells. J Thromb Haemost 2006; 4:217-28. [PMID: 16409472 DOI: 10.1111/j.1538-7836.2005.01654.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The molecular defect of a new Bernard-Soulier patient, originating from Morocco and presenting thrombocytopenia with large platelets and an absence of ristocetin-induced platelet agglutination, has been identified and reproduced in transfected heterologous cells. Gene sequencing revealed insertion of a guanine in the domain coding for the transmembrane region of the glycoprotein (GP) Ib beta subunit. This mutation causes a translational frame shift, which creates putative novel transmembrane and cytoplasmic 37 and 125 amino acids domains, respectively. A 34 kDa immunoreactive GPIb beta band, instead of the normal 26 kDa subunit, was detected by Western blotting in lysates from the patient's platelets and from transfected cells and in immunoprecipitates of metabolically labeled cells. The abnormal subunit did not associate with GPIb alpha and was mainly intracellular, although a significant fraction could reach the cell surface. Cells expressing the mutant GPIb-IX complex adhered to a von Willebrand factor matrix but were unable to change shape, unlike cells expressing the wild-type receptor. These results strongly suggest a novel role of the GPIb beta subunit and its transmembrane-intracellular region in GPIb-VWF-dependent signaling, in addition to a role in correct assembly and cell surface targeting of the GPIb-V-IX complex.
Collapse
Affiliation(s)
- C Strassel
- INSERM U.311, Etablissement Français du Sang, Alsace, Strasbourg, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Viral rescue of murine Glanzmann thrombasthenia. Blood 2005. [DOI: 10.1182/blood-2005-07-3061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
39
|
Abstract
Platelets play a central role in hemostasis and thrombosis but also in the initiation of atherosclerosis, making platelet receptors and their intracellular signaling pathways important molecular targets for antithrombotic and anti-inflammatory therapy. Historically, much of the knowledge about hemostasis and thrombosis has been derived from patients suffering from bleeding and thrombotic disorders and the identification of the underlying molecular defects. In recent years, the availability of genetically modified mouse strains with defined defects in platelet function and the development of in vivo models to assess platelet-related physiologic and pathophysiologic processes have opened new ways to identify the individual roles and the interplay of platelet proteins in adhesion, activation, aggregation, secretion, and procoagulant activity in vitro and in vivo. This review will summarize key findings made by these approaches and discuss them in the context of human disease.
Collapse
Affiliation(s)
- B Nieswandt
- Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany.
| | | | | | | |
Collapse
|