151
|
Pierre S, Linke B, Suo J, Tarighi N, Del Turco D, Thomas D, Ferreiros N, Stegner D, Frölich S, Sisignano M, Meyer Dos Santos S, deBruin N, Nüsing RM, Deller T, Nieswandt B, Geisslinger G, Scholich K. GPVI and Thromboxane Receptor on Platelets Promote Proinflammatory Macrophage Phenotypes during Cutaneous Inflammation. J Invest Dermatol 2016; 137:686-695. [PMID: 27818280 DOI: 10.1016/j.jid.2016.09.036] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 09/13/2016] [Indexed: 01/07/2023]
Abstract
Platelets are well known for their role in hemostasis but are also increasingly recognized for their supporting role in innate immune responses. Here, we studied the role of platelets in the development of peripheral inflammation and found that platelets colocalize with macrophages in the inflamed tissue outside of blood vessels in different animal models for cutaneous inflammation. Collagen-treatment of macrophages isolated from paws during zymosan-induced inflammation induced thromboxane synthesis through the platelet-expressed collagen receptor glycoprotein VI. Deletion of glycoprotein VI or its downstream effector thromboxane A2 receptor (TP) reduced zymosan-induced mechanical allodynia without altering macrophage recruitment or formation of macrophage/platelet complexes. Instead, macrophages in inflamed paws of glycoprotein VI- and TP-deficient mice exhibited an increased expression of anti-inflammatory markers and synthesized less proinflammatory mediators (prostaglandin E2 and IL6). TP expression on platelets was necessary to mediate increased prostaglandin E2 and IL6 synthesis, whereas TP expression on macrophages was sufficient to decrease the expression of the anti-inflammatory macrophage marker CD206, showing that TP activation on platelets and macrophages regulates different aspects of macrophage activation.
Collapse
Affiliation(s)
- Sandra Pierre
- Institut für Klinische Pharmakologie, pharmazentrum frankfurt, ZAFES, Klinikum der Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Bona Linke
- Institut für Klinische Pharmakologie, pharmazentrum frankfurt, ZAFES, Klinikum der Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Jing Suo
- Institut für Klinische Pharmakologie, pharmazentrum frankfurt, ZAFES, Klinikum der Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Neda Tarighi
- Institut für Klinische Pharmakologie, pharmazentrum frankfurt, ZAFES, Klinikum der Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Domenico Del Turco
- Institut für Klinische Neuroanatomie, Neuroscience Center, Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Dominique Thomas
- Institut für Klinische Pharmakologie, pharmazentrum frankfurt, ZAFES, Klinikum der Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Nerea Ferreiros
- Institut für Klinische Pharmakologie, pharmazentrum frankfurt, ZAFES, Klinikum der Goethe-Universität Frankfurt, Frankfurt, Germany
| | - David Stegner
- Universität Würzburg, Institut für Experimentelle Biomedizin, Universitätsklinikum und Rudolf-Virchow-Zentrum für Experimentelle Biomedizin, Würzburg, Germany
| | - Stefanie Frölich
- Institut für Klinische Pharmakologie, pharmazentrum frankfurt, ZAFES, Klinikum der Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Marco Sisignano
- Institut für Klinische Pharmakologie, pharmazentrum frankfurt, ZAFES, Klinikum der Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Sascha Meyer Dos Santos
- Institut für Klinische Pharmakologie, pharmazentrum frankfurt, ZAFES, Klinikum der Goethe-Universität Frankfurt, Frankfurt, Germany; Fraunhofer Institute of Molecular Biology and Applied Ecology-Project Group Translational Medicine and Pharmacology (IME-TMP), Frankfurt, Germany
| | - Natasja deBruin
- Fraunhofer Institute of Molecular Biology and Applied Ecology-Project Group Translational Medicine and Pharmacology (IME-TMP), Frankfurt, Germany
| | - Rolf M Nüsing
- Institut für Klinische Pharmakologie, pharmazentrum frankfurt, ZAFES, Klinikum der Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Thomas Deller
- Institut für Klinische Neuroanatomie, Neuroscience Center, Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Bernhard Nieswandt
- Universität Würzburg, Institut für Experimentelle Biomedizin, Universitätsklinikum und Rudolf-Virchow-Zentrum für Experimentelle Biomedizin, Würzburg, Germany
| | - Gerd Geisslinger
- Institut für Klinische Pharmakologie, pharmazentrum frankfurt, ZAFES, Klinikum der Goethe-Universität Frankfurt, Frankfurt, Germany; Fraunhofer Institute of Molecular Biology and Applied Ecology-Project Group Translational Medicine and Pharmacology (IME-TMP), Frankfurt, Germany
| | - Klaus Scholich
- Institut für Klinische Pharmakologie, pharmazentrum frankfurt, ZAFES, Klinikum der Goethe-Universität Frankfurt, Frankfurt, Germany.
| |
Collapse
|
152
|
Chatterjee M, Gawaz M. Clinical significance of receptor shedding-platelet GPVI as an emerging diagnostic and therapeutic tool. Platelets 2016; 28:362-371. [PMID: 27753514 DOI: 10.1080/09537104.2016.1227062] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Platelet membrane bedecked with a wide array of receptors offers a platform to regulate platelet responsiveness, thrombotic propensity, inflammatory disposition, and immune reactivity under diverse pathophysiological conditions. Ectopic proteolytic cleavage of such receptors irreversibly inactivates receptor-mediated intracellular signaling governing cellular functions, further releases soluble fragments into circulation which might modulate functions of target cells. Glycoprotein VI-(GPVI) is a membrane glycoprotein expressed in platelets and megakaryocytes. Platelet GPVI surface expression is enhanced following acute ischemic events like myocardial infarction and cerebral stroke, serves as an imminent diagnostic tool independent of markers of tissue necrosis, and is associated with poor prognosis. Platelets undergo GPVI shedding and thereby contribute to soluble plasma levels of sGPVI, with distinct diagnostic and prognostic attributes. This review summarizes the functional significance and mechanistic basis whereby GPVI surface availability is up- or downregulated on platelets and the impact of GPVI in diagnostic, prognostic, and therapeutic strategies in diseases where platelets play a regulatory role. Further, we also highlight how novel non-invasive platelet-based diagnostic and therapeutic strategies have evolved utilizing GPVI for lesion-directed antithrombotic therapy or to counteract atherosclerotic disposition to ameliorate care of patients particularly in the context of cardio-cerebro-vascular medicine.
Collapse
Affiliation(s)
- Madhumita Chatterjee
- a Innere Medizin III, Kardiologie und Kreislauferkrankungen , Eberhard Karls Universität , Tübingen , Germany
| | - Meinrad Gawaz
- a Innere Medizin III, Kardiologie und Kreislauferkrankungen , Eberhard Karls Universität , Tübingen , Germany
| |
Collapse
|
153
|
Determination of structural requirements of Mer kinase inhibitors and binding interaction analysis using in silico approaches. Med Chem Res 2016. [DOI: 10.1007/s00044-016-1722-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
154
|
Affiliation(s)
- Markus Bender
- Department of Experimental Biomedicine, University of Würzburg, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| | - David Stegner
- Department of Experimental Biomedicine, University of Würzburg, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| | - Bernhard Nieswandt
- Department of Experimental Biomedicine, University of Würzburg, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| |
Collapse
|
155
|
Cevik O, Baykal AT, Sener A. Platelets Proteomic Profiles of Acute Ischemic Stroke Patients. PLoS One 2016; 11:e0158287. [PMID: 27336623 PMCID: PMC4919045 DOI: 10.1371/journal.pone.0158287] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 06/13/2016] [Indexed: 12/20/2022] Open
Abstract
Platelets play a crucial role in the pathogenesis of stroke and antiplatelet agents exist for its treatment and prevention. Through the use of LC-MS based protein expression profiling, platelets from stroke patients were analyzed and then correlated with the proteomic analyses results in the context of this disease. This study was based on patients who post ischemic stroke were admitted to hospital and had venous blood drawn within 24 hrs of the incidence. Label-free protein expression analyses of the platelets' tryptic digest was performed in triplicate on a UPLC-ESI-qTOF-MS/MS system and ProteinLynx Global Server (v2.5, Waters) was used for tandem mass data extraction. The peptide sequences were searched against the reviewed homo sapiens database (www.uniprot.org) and the quantitation of protein variation was achieved through Progenesis LC-MS software (V4.0, Nonlinear Dynamics). These Label-free differential proteomics analysis of platelets ensured that 500 proteins were identified and 83 of these proteins were found to be statistically significant. The differentially expressed proteins are involved in various processes such as inflammatory response, cellular movement, immune cell trafficking, cell-to-cell signaling and interaction, hematological system development and function and nucleic acid metabolism. The expressions of myeloperoxidase, arachidonate 12-Lipoxygenase and histidine-rich glycoprotein are involved in cellular metabolic processes, crk-like protein and ras homolog gene family member A involved in cell signaling with vitronectin, thrombospondin 1, Integrin alpha 2b, and integrin beta 3 involved in cell adhesion. Apolipoprotein H, immunoglobulin heavy constant gamma 1 and immunoglobulin heavy constant gamma 3 are involved in structural, apolipoprotein A-I, and alpha-1-microglobulin/bikunin precursor is involved in transport, complement component 3 and clusterin is involved in immunity proteins as has been discussed. Our data provides an insight into the proteins that are involved in the platelets' activation response during ischemic stroke. It could be argued that this study lays the foundation for future mechanistic studies.
Collapse
Affiliation(s)
- Ozge Cevik
- Cumhuriyet University, Faculty of Pharmacy, Department of Biochemistry, Sivas, Turkey
- Marmara University, Faculty of Pharmacy, Department of Biochemistry, Istanbul, Turkey
- * E-mail:
| | - Ahmet Tarik Baykal
- Acibadem University, School of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
| | - Azize Sener
- Marmara University, Faculty of Pharmacy, Department of Biochemistry, Istanbul, Turkey
| |
Collapse
|
156
|
Induruwa I, Jung SM, Warburton EA. Beyond antiplatelets: The role of glycoprotein VI in ischemic stroke. Int J Stroke 2016; 11:618-25. [PMID: 27312676 PMCID: PMC5390959 DOI: 10.1177/1747493016654532] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/03/2016] [Indexed: 11/17/2022]
Abstract
Background Platelets are essential to physiological hemostasis or pathological thrombus formation. Current antiplatelet agents inhibit platelet aggregation but leave patients at risk of systemic side-effects such as hemorrhage. Newer therapeutic strategies could involve targeting this cascade earlier during platelet adhesion or activation via inhibitory effects on specific glycoproteins, the thrombogenic collagen receptors found on the platelet surface. Aims Glycoprotein VI (GPVI) is increasingly being recognized as the main platelet-collagen receptor involved in arterial thrombosis. This review summarizes the crucial role GPVI plays in ischemic stroke as well as the current strategies used to attempt to inhibit its activity. Summary of review In this review, we discuss the normal hemostatic process, and the role GPVI plays at sites of atherosclerotic plaque rupture. We discuss how the unique structure of GPVI allows for its interaction with collagen and creates downstream signaling that leads to thrombus formation. We summarize the current strategies used to inhibit GPVI activity and how this could translate to a clinically viable entity that may compete with current antiplatelet therapy. Conclusion From animal models, it is clear that GPVI inhibition leads to an abolished platelet response to collagen and reduced platelet aggregation, culminating in smaller arterial thrombi. There is now an increasing body of evidence that these findings can be translated into the development of a bleeding free pharmacological entity specific to sites of plaque rupture in humans.
Collapse
Affiliation(s)
- Isuru Induruwa
- Department of Clinical Neurosciences, Box 83, Cambridge University Biomedical Campus, Cambridge, UK
| | - Stephanie M Jung
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Elizabeth A Warburton
- Department of Clinical Neurosciences, Box 83, Cambridge University Biomedical Campus, Cambridge, UK
| |
Collapse
|
157
|
Yun SH, Sim EH, Goh RY, Park JI, Han JY. Platelet Activation: The Mechanisms and Potential Biomarkers. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9060143. [PMID: 27403440 PMCID: PMC4925965 DOI: 10.1155/2016/9060143] [Citation(s) in RCA: 319] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 05/26/2016] [Indexed: 11/17/2022]
Abstract
Beyond hemostasis and thrombosis, an increasing number of studies indicate that platelets play an integral role in intercellular communication, mediating inflammatory and immunomodulatory activities. Our knowledge about how platelets modulate inflammatory and immunity has greatly improved in recent years. In this review, we discuss recent advances in the pathways of platelet activation and potential application of platelet activation biomarkers to diagnosis and prediction of disease states.
Collapse
Affiliation(s)
- Seong-Hoon Yun
- Department of Biochemistry, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan 49201, Republic of Korea
| | - Eun-Hye Sim
- Department of Biochemistry, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan 49201, Republic of Korea
| | - Ri-Young Goh
- Department of Laboratory Medicine, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan 49201, Republic of Korea
| | - Joo-In Park
- Department of Biochemistry, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan 49201, Republic of Korea
| | - Jin-Yeong Han
- Department of Laboratory Medicine, Dong-A University College of Medicine, 26 Daesingongwon-ro, Seo-gu, Busan 49201, Republic of Korea
| |
Collapse
|
158
|
Milanowski L, Pordzik J, Janicki PK, Postula M. Common genetic variants in platelet surface receptors and its association with ischemic stroke. Pharmacogenomics 2016; 17:953-71. [PMID: 27269246 DOI: 10.2217/pgs.16.21] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Ischemic stroke has been named one of the leading causes of death worldwide. Whereas numerous biological mechanisms and molecules were found to be associated with stroke, platelets are particularly contributive to its pathogenesis. Recent data indicate considerable variability in platelet phenotype which accounts for differences in platelet surface receptor function, count and reactivity. These features collectively influence both the events leading to a disease and effectiveness of antiplatelet therapies. Consequently, genetic variants predisposing to cerebrovascular diseases can be sequenced using a wide array of techniques and become a useful tool in clinical setting. In this review, we provide an outline of common platelet polymorphisms that impose risk on ischemic stroke development and should be evaluated as targets to improve treatment. As study results are often inconsistent, partly due to differences in demographic characteristics between study populations and the fact that the functional impact of these variants has been relatively small, we conclude that both rare, low-frequency and common variants might account for genetic contribution on abnormal platelet response to antiplatelet drugs.
Collapse
Affiliation(s)
- Lukasz Milanowski
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
| | - Justyna Pordzik
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland
| | - Piotr K Janicki
- Perioperative Genomics Laboratory, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Center for Preclinical Research and Technology CEPT, Warsaw, Poland.,Perioperative Genomics Laboratory, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
159
|
Saito MS, Lourenço AL, Kang HC, Rodrigues CR, Cabral LM, Castro HC, Satlher PC. New approaches in tail-bleeding assay in mice: improving an important method for designing new anti-thrombotic agents. Int J Exp Pathol 2016; 97:285-92. [PMID: 27377432 PMCID: PMC4960579 DOI: 10.1111/iep.12182] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 03/10/2016] [Indexed: 12/11/2022] Open
Abstract
This report describes a modified, simple, low-cost and more sensitive method to determine bleeding patterns and haemoglobin concentration in a tail-bleeding assay using BALB/c mice and tail tip amputation. The cut tail was immersed in Drabkin's reagent to promote erythrocyte lysis and haemoglobin release, which was monitored over 30 min. The operator was blinded to individual conditions of the mice, which were treated with either saline (NaCl 0.15m), DMSO (0.5%) or clinical anti-thrombotic drugs. Our experimental protocols showed good reproducibility and repeatability of results when using Drabkin's reagent than water. Thus, the use of Drabkin's reagent offered a simple and low-cost method to observe and quantify the bleeding and rebleeding episodes. We also observed the bleeding pattern and total haemoglobin loss using untreated animals or those under anti-coagulant therapy in order to validate the new Drabkin method and thus confirm that it is a useful protocol to quantify haemoglobin concentrations in tail-bleeding assay. This modified method provided a more accurate results for bleeding patterns in mice and for identifying new anti-thrombotic drugs.
Collapse
Affiliation(s)
- Max Seidy Saito
- Laboratório de Antibióticos Bioquímica Ensino e Modelagem Molecular (LABiEMol) - Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
- Programa de Pós-Graduação em Patologia (PPG-UFF) - Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - André Luiz Lourenço
- Laboratório de Antibióticos Bioquímica Ensino e Modelagem Molecular (LABiEMol) - Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
- Programa de Pós-Graduação em Patologia (PPG-UFF) - Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Hye Chung Kang
- Programa de Pós-Graduação em Patologia (PPG-UFF) - Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Carlos Rangel Rodrigues
- Laboratório de Modelagem Molecular e QSAR (ModMolQSAR) - Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucio Mendes Cabral
- Laboratório de Tecnologia Industrial Farmacêutica (LabTIF) - Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Helena Carla Castro
- Laboratório de Antibióticos Bioquímica Ensino e Modelagem Molecular (LABiEMol) - Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
| | - Plínio Cunha Satlher
- Laboratório de Antibióticos Bioquímica Ensino e Modelagem Molecular (LABiEMol) - Instituto de Biologia, Universidade Federal Fluminense, Niterói, RJ, Brazil
- Laboratório de Tecnologia Industrial Farmacêutica (LabTIF) - Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
160
|
Development of antithrombotic nanoconjugate blocking integrin α2β1-collagen interactions. Sci Rep 2016; 6:26292. [PMID: 27195826 PMCID: PMC4872532 DOI: 10.1038/srep26292] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/28/2016] [Indexed: 01/07/2023] Open
Abstract
An antithrombotic nanoconjugate was designed in which a designed biomimetic peptide LWWNSYY was immobilized to the surface of poly(glycidyl methacrylate) nanoparticles (PGMA NPs). Our previous work has demonstrated LWWNSYY to be an effective inhibitor of integrin α2β1-collagen interaction and subsequent thrombus formation, however its practical application suffered from the formation of clusters in physiological environment caused by its high hydrophobicity. In our present study, the obtained LWWNSYY-PGMA nanoparticles (L-PGMA NPs) conjugate, with an improved dispersibility of LWWNSYY by PGMA NPs, have shown binding to collagen receptors with a Kd of 3.45 ± 1.06 μM. L-PGMA NPs have also proven capable of inhibiting platelet adhesion in vitro with a reduced IC50 of 1.83 ± 0.29 μg/mL. High inhibition efficiency of L-PGMA NPs in thrombus formation was further confirmed in vivo with a 50% reduction of thrombus weight. Therefore, L-PGMA NPs were developed as a high-efficiency antithrombotic nanomedicine targeted for collagen exposed on diseased blood vessel wall.
Collapse
|
161
|
Vaddi K, Verstovsek S, Kiladjian JJ. Ruxolitinib: a targeted treatment option for patients with polycythemia vera. BLOOD AND LYMPHATIC CANCER-TARGETS AND THERAPY 2016; 6:7-19. [PMID: 31360077 PMCID: PMC6467337 DOI: 10.2147/blctt.s101185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Polycythemia vera (PV) is a chronic myeloproliferative neoplasm characterized by erythrocytosis and the presence of Janus kinase (JAK) 2V617F or similar mutations. This review summarizes the pathophysiology of PV, the challenges associated with traditional treatment options, and the scientific rationale and supportive clinical evidence for targeted therapy with ruxolitinib. Accumulating evidence indicates that activating mutations in JAK2 drive the PV disease state. Traditional PV treatment strategies, including aspirin, phlebotomy, and cytoreductive agents such as hydroxyurea, provide clinical benefits for some but not all patients and may not adequately treat PV-related symptoms. Furthermore, traditional treatment approaches are associated with potential side effects that may limit their usage and lead some patients to discontinue the treatment. Ruxolitinib is an orally available small-molecule tyrosine kinase inhibitor that is a potent and selective inhibitor of JAK1/JAK2. Ruxolitinib is approved in the US for patients with PV with an inadequate response or intolerance to hydroxyurea and in Europe for adults with PV who are resistant to or intolerant of hydroxyurea. In the Phase III RESPONSE registration trial, ruxolitinib was superior to the best available therapy in patients with PV who were resistant to or intolerant of hydroxyurea in controlling hematocrit levels, reducing spleen volume, and improving PV-related symptoms and quality-of-life measures. The most common nonhematologic adverse events in ruxolitinib-treated patients were headache, diarrhea, pruritus, and fatigue in the RESPONSE trial; hematologic adverse events were primarily grade 1 or 2. In the Phase IIIb nonregistration RELIEF trial, there were nonsignificant trends toward an improved symptom control in patients with PV on a stable hydroxyurea dose who were generally well controlled but reported disease-associated symptoms and switched to ruxolitinib vs those who continued hydroxyurea therapy. Updated treatment guidelines will be important for educating physicians about the role of ruxolitinib in the treatment of patients with PV.
Collapse
Affiliation(s)
- Kris Vaddi
- Drug Discovery, Incyte Corporation, Wilmington, DE,
| | - Srdan Verstovsek
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jean-Jacques Kiladjian
- Clinical Investigations Center, Hôpital Saint-Louis et Université Paris Diderot, Paris, France
| |
Collapse
|
162
|
Nguyen TH, Palankar R, Bui VC, Medvedev N, Greinacher A, Delcea M. Rupture Forces among Human Blood Platelets at different Degrees of Activation. Sci Rep 2016; 6:25402. [PMID: 27146004 PMCID: PMC4857101 DOI: 10.1038/srep25402] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/18/2016] [Indexed: 11/21/2022] Open
Abstract
Little is known about mechanics underlying the interaction among platelets during activation and aggregation. Although the strength of a blood thrombus has likely major biological importance, no previous study has measured directly the adhesion forces of single platelet-platelet interaction at different activation states. Here, we filled this void first, by minimizing surface mediated platelet-activation and second, by generating a strong adhesion force between a single platelet and an AFM cantilever, preventing early platelet detachment. We applied our setup to measure rupture forces between two platelets using different platelet activation states, and blockade of platelet receptors. The rupture force was found to increase proportionally to the degree of platelet activation, but reduced with blockade of specific platelet receptors. Quantification of single platelet-platelet interaction provides major perspectives for testing and improving biocompatibility of new materials; quantifying the effect of drugs on platelet function; and assessing the mechanical characteristics of acquired/inherited platelet defects.
Collapse
Affiliation(s)
- Thi-Huong Nguyen
- Nanostructure Group, ZIK HIKE - Center for Innovation Competence, Humoral Immune Reactions in Cardiovascular Diseases, University of Greifswald, 17489 Greifswald, Germany.,Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Raghavendra Palankar
- Nanostructure Group, ZIK HIKE - Center for Innovation Competence, Humoral Immune Reactions in Cardiovascular Diseases, University of Greifswald, 17489 Greifswald, Germany.,Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Van-Chien Bui
- Nanostructure Group, ZIK HIKE - Center for Innovation Competence, Humoral Immune Reactions in Cardiovascular Diseases, University of Greifswald, 17489 Greifswald, Germany.,Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Nikolay Medvedev
- Nanostructure Group, ZIK HIKE - Center for Innovation Competence, Humoral Immune Reactions in Cardiovascular Diseases, University of Greifswald, 17489 Greifswald, Germany
| | - Andreas Greinacher
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Mihaela Delcea
- Nanostructure Group, ZIK HIKE - Center for Innovation Competence, Humoral Immune Reactions in Cardiovascular Diseases, University of Greifswald, 17489 Greifswald, Germany.,Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| |
Collapse
|
163
|
El Haouari M, Rosado JA. Medicinal Plants with Antiplatelet Activity. Phytother Res 2016; 30:1059-71. [PMID: 27062716 DOI: 10.1002/ptr.5619] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 02/29/2016] [Accepted: 03/12/2016] [Indexed: 12/25/2022]
Abstract
Blood platelets play an essential role in the hemostasis and wound-healing processes. However, platelet hyperactivity is associated to the development and the complications of several cardiovascular diseases. In this sense, the search for potent and safer antiplatelet agents is of great interest. This article provides an overview of experimental studies performed on medicinal plants with antiplatelet activity available through literature with particular emphasis on the bioactive constituents, the parts used, and the various platelet signaling pathways modulated by medicinal plants. From this review, it was suggested that medicinal plants with antiplatelet activity mainly belong to the family of Asteraceae, Rutaceae, Fabaceae, Lamiaceae, Zygophyllaceae, Rhamnaceae, Liliaceae, and Zingiberaceae. The antiplatelet effect is attributed to the presence of bioactive compounds such as polyphenols, flavonoids, coumarins, terpenoids, and other substances which correct platelet abnormalities by interfering with different platelet signalization pathways including inhibition of the ADP pathway, suppression of TXA2 formation, reduction of intracellular Ca(2+) mobilization, and phosphoinositide breakdown, among others. The identification and/or structure modification of the plant constituents and the understanding of their action mechanisms will be helpful in the development of new antiplatelet agents based on medicinal plants which could contribute to the prevention of thromboembolic-related disorders by inhibiting platelet aggregation. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Mohammed El Haouari
- Centre Régional des Métiers de l'Education et de la Formation de Taza (CRMEF - Taza), B.P. 1178, Taza Gare, Morocco.,Faculté Polydisciplinaire de Taza, Laboratoire des Matériaux, Substances Naturelles, Environnement et Modélisation (LMSNEM), Université Sidi Mohamed Ben Abdellah, B.P. 1223, Taza Gare, Morocco
| | - Juan A Rosado
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003, Cáceres, Spain
| |
Collapse
|
164
|
Schmidt EP, Kuebler WM, Lee WL, Downey GP. Adhesion Molecules: Master Controllers of the Circulatory System. Compr Physiol 2016; 6:945-73. [PMID: 27065171 DOI: 10.1002/cphy.c150020] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This manuscript will review our current understanding of cellular adhesion molecules (CAMs) relevant to the circulatory system, their physiological role in control of vascular homeostasis, innate and adaptive immune responses, and their importance in pathophysiological (disease) processes such as acute lung injury, atherosclerosis, and pulmonary hypertension. This is a complex and rapidly changing area of research that is incompletely understood. By design, we will begin with a brief overview of the structure and classification of the major groups of adhesion molecules and their physiological functions including cellular adhesion and signaling. The role of specific CAMs in the process of platelet aggregation and hemostasis and leukocyte adhesion and transendothelial migration will be reviewed as examples of the complex and cooperative interplay between CAMs during physiological and pathophysiological processes. The role of the endothelial glycocalyx and the glycobiology of this complex system related to inflammatory states such as sepsis will be reviewed. We will then focus on the role of adhesion molecules in the pathogenesis of specific disease processes involving the lungs and cardiovascular system. The potential of targeting adhesion molecules in the treatment of immune and inflammatory diseases will be highlighted in the relevant sections throughout the manuscript.
Collapse
Affiliation(s)
- Eric P Schmidt
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Wolfgang M Kuebler
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Departments of Surgery and Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Warren L Lee
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Division of Respirology and the Interdepartmental Division of Critical Care Medicine, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Gregory P Downey
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Departments of Medicine, Pediatrics, and Biomedical Research, National Jewish Health, Denver, Colorado, USA
- Departments of Medicine, and Immunology and Microbiology, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
165
|
Rekittke NE, Ang M, Rawat D, Khatri R, Linn T. Regenerative Therapy of Type 1 Diabetes Mellitus: From Pancreatic Islet Transplantation to Mesenchymal Stem Cells. Stem Cells Int 2016; 2016:3764681. [PMID: 27047547 PMCID: PMC4800095 DOI: 10.1155/2016/3764681] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/14/2015] [Indexed: 01/01/2023] Open
Abstract
Type 1 diabetes is an autoimmune disease resulting in the permanent destruction of pancreatic islets. Islet transplantation to portal vein provides an approach to compensate for loss of insulin producing cells. Clinical trials demonstrated that even partial islet graft function reduces severe hypoglycemic events in patients. However, therapeutic impact is restrained due to shortage of pancreas organ donors and instant inflammation occurring in the hepatic environment of the graft. We summarize on what is known about regenerative therapy in type 1 diabetes focusing on pancreatic islet transplantation and new avenues of cell substitution. Metabolic pathways and energy production of transplanted cells are required to be balanced and protection from inflammation in their intravascular bed is desired. Mesenchymal stem cells (MSCs) have anti-inflammatory features, and so they are interesting as a therapy for type 1 diabetes. Recently, they were reported to reduce hyperglycemia in diabetic rodents, and they were even discussed as being turned into endodermal or pancreatic progenitor cells. MSCs are recognized to meet the demand of an individual therapy not raising the concerns of embryonic or induced pluripotent stem cells for therapy.
Collapse
Affiliation(s)
- Nadine E. Rekittke
- Clinical Research Unit, Zentrum für Innere Medizin, Fachbereich Medizin, Justus Liebig Universität Giessen, 35392 Giessen, Germany
| | - Meidjie Ang
- Clinical Research Unit, Zentrum für Innere Medizin, Fachbereich Medizin, Justus Liebig Universität Giessen, 35392 Giessen, Germany
| | - Divya Rawat
- Clinical Research Unit, Zentrum für Innere Medizin, Fachbereich Medizin, Justus Liebig Universität Giessen, 35392 Giessen, Germany
| | - Rahul Khatri
- Clinical Research Unit, Zentrum für Innere Medizin, Fachbereich Medizin, Justus Liebig Universität Giessen, 35392 Giessen, Germany
| | - Thomas Linn
- Clinical Research Unit, Zentrum für Innere Medizin, Fachbereich Medizin, Justus Liebig Universität Giessen, 35392 Giessen, Germany
| |
Collapse
|
166
|
Moudgil R, Yeh ETH. Mechanisms of Cardiotoxicity of Cancer Chemotherapeutic Agents: Cardiomyopathy and Beyond. Can J Cardiol 2016; 32:863-870.e5. [PMID: 27117975 DOI: 10.1016/j.cjca.2016.01.027] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/26/2016] [Accepted: 01/27/2016] [Indexed: 12/14/2022] Open
Abstract
Tremendous strides have been made in the treatment of various oncological diseases such that patients are surviving longer and are having better quality of life. However, the success has been tainted by the iatrogenic cardiac toxicities. This is especially concerning in the younger population who are facing cardiac disease such as heart failure in their 30s and 40s as the consequence of the anthracycline's side effects (used for childhood leukemia and lymphoma). This resulted in the awareness of cardiotoxic effects of anticancer drugs and emergence of a new discipline: oncocardiology. Since then, numerous anticancer drugs have been correlated to cardiomyopathy. Additionally, other cardiovascular effects have been identified, which includes but is not limited to myocardial infarction, thrombosis, hypertension, arrhythmias, and pulmonary hypertension. In this review we examine some of the anticancer agents that mitigate cardiotoxicity and present current knowledge of molecular mechanism(s). The aim of the review is to ignite awareness of emerging cardiotoxic effects as new generations of anticancer agents are being tested in clinical trials and introduced as part of the therapeutic armamentarium to our oncological patients.
Collapse
Affiliation(s)
- Rohit Moudgil
- Department of Cardiology, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Edward T H Yeh
- Department of Cardiology, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
167
|
Burchell SR, Iniaghe LO, Zhang JH, Tang J. Fucoidan from Fucus vesiculosus Fails to Improve Outcomes Following Intracerebral Hemorrhage in Mice. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:191-8. [PMID: 26463947 DOI: 10.1007/978-3-319-18497-5_34] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Intracerebral hemorrhage (ICH) is the most fatal stroke subtype, with no effective therapies. Hematoma expansion and inflammation play major roles in the pathophysiology of ICH, contributing to primary and secondary brain injury, respectively. Fucoidan, a polysaccharide from the brown seaweed Fucus vesiculosus, has been reported to activate a platelet receptor that may function in limiting bleeding, and to exhibit anti-inflammatory effects. As such, the aim of the present study was to examine the effects of fucoidan on hemorrhaging and neurological outcomes after ICH. Male CD-1 mice were subjected to experimental ICH by infusion of bacterial collagenase. Animals were randomly divided into the following groups: sham, ICH + vehicle, ICH + 25 mg/kg fucoidan, ICH + 75 mg/kg fucoidan, and ICH + 100 mg/kg fucoidan. Brain water content, neurobehavioral outcomes, and hemoglobin content were evaluated at 24 h post ICH. Our findings show that fucoidan failed to attenuate the ICH-induced increase in BWC. The neurological deficits that result from ICH also did not differ in the treatment groups at all three doses. Finally, we found that fucoidan had no effect on the hemoglobin content after ICH. We postulate that fucoidan treatment did not improve the measured outcomes after ICH because we used crude fucoidan, which has a high molecular weight, in our study. High-molecular-weight fucoidans are reported to have less therapeutic potential than low molecular weight fucoidans. They have been shown to exhibit anti-coagulant and pro-apoptotic properties, which seem to outweigh their anti-inflammatory and potential procoagulant abilities. We propose that using a low-molecular-weight fucoidan, or fractionating the crude polysaccharide, may be effective in treating ICH. Future studies are needed to confirm this.
Collapse
Affiliation(s)
- Sherrefa R Burchell
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Loretta O Iniaghe
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
- Department of Pharmacology and Toxicology, University of Benin, Benin City, Nigeria
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| |
Collapse
|
168
|
de Villiers S, Swanepoel A, Bester J, Pretorius E. Novel Diagnostic and Monitoring Tools in Stroke: an Individualized Patient-Centered Precision Medicine Approach. J Atheroscler Thromb 2015; 23:493-504. [PMID: 26686739 DOI: 10.5551/jat.32748] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Central to the pathogenesis of ischaemic stroke are the normally protective processes of platelet adhesion and activation. Experimental evidence has shown that the ligand-receptor interactions in ischaemic stroke represent a thrombo-inflammatory cascade, which presents research opportunities into new treatment. However, as anti-platelet drugs have the potential to cause severe side effects in ischaemic stroke patients (as well as other vascular disease patients), it is important to carefully monitor the risk of bleeding and risk of thrombus in patients receiving treatment. Because thrombo-embolic ischaemic stroke is a major health issue, we suggest that the answer to adequate treatment is based on an individualized patient-centered approach, inline with the latest NIH precision medicine approach. A combination of viscoelastic methodologies may be used in a personalized patient-centered regime, including thromboelastography (TEG®) and the lesser used scanning electron microscopy approach (SEM). Thromboelastography provides a dynamic measure of clot formation, strength, and lysis, whereas SEM is a visual structural tool to study patient fibrin structure in great detail. Therefore, we consider the evidence for TEG® and SEM as unique means to confirm stroke diagnosis, screen at-risk patients, and monitor treatment efficacy. Here we argue that the current approach to stroke treatment needs to be restructured and new innovative thought patterns need to be applied, as even approved therapies require close patient monitoring to determine efficacy, match treatment regimens to each patient's individual needs, and assess the risk of dangerous adverse effects. TEG® and SEM have the potential to be a useful tool and could potentially alter the clinical approach to managing ischaemic stroke. As envisaged in the NIH precision medicine approach, this will involve a number of role players and innovative new research ideas, with benefits that will ultimately only be realized in a few years. Therefore, with this ultimate goal in mind, we suggest that an individualized patient-orientated approach is now available and therefore already within our ability to use.
Collapse
Affiliation(s)
- Sulette de Villiers
- Department of Physiology, Faculty of Health Sciences, University of Pretoria
| | | | | | | |
Collapse
|
169
|
Chiou TJ, Chang YF, Wang MC, Kao CW, Lin HY, Chen TY, Hsueh EJ, Lan YJ, Sung YC, Lin SF, Bai LY, Chen CG. Eltrombopag enhances platelet adhesion by upregulating the expression of glycoprotein VI in patients with chronic immune thrombocytopenic purpura. Transl Res 2015; 166:750-761.e4. [PMID: 26477577 DOI: 10.1016/j.trsl.2015.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/13/2015] [Accepted: 09/14/2015] [Indexed: 10/23/2022]
Abstract
Eltrombopag, a thrombopoietin receptor agonist, has been approved for the treatment of patients with immune thrombocytopenia because of its abilities to enhance platelet production and reduce hemorrhage. Both platelet count and platelet adhesion are crucial to stop bleeding. Although eltrombopag is known to improve platelet counts, its effects on platelet adhesion are not yet known. This study aimed to assess the efficacy of eltrombopag on platelet production and platelet adhesive affinity. To evaluate the efficacy of low-dose eltrombopag (25 mg) for patients with chronic refractory immune thrombocytopenic purpura (ITP) and to determine the ex vivo platelet adhesion ability before and after treatment with eltrombopag, we conducted an open-label, multicenter study in which 25 Taiwanese patients with chronic ITP were enrolled. During the 6-month evaluation, the starting and maximum doses of eltrombopag were 25 and 50 mg, respectively, to maintain the platelet count of ≥50,000 per μL. Flow-based adhesion assay was used to detect the percentage of platelets adhering to immobilized von Willebrand factor-collagen on microslides. Of the enrolled patients, 48% achieved a platelet count of ≥50,000 per μL. Interestingly, 83% of all responders required 25 mg of eltrombopag daily to achieve the target platelet count. In addition, the percentage of bleeding patients was significantly reduced in both responders and nonresponders by 50% from the baseline level throughout the treatment period. The ex vivo platelet adhesion capacity was elevated after the 6-month eltrombopag treatment in both responders and nonresponders. Furthermore, glycoprotein VI (GPVI) expression was significantly upregulated after treatment with eltrombopag. Low-to-intermediate dose of eltrombopag showed good efficacy to expedite platelet production and augment platelet adhesion. These 2 factors might explain the efficacy of eltrombopag in ameliorating hemorrhage in patients with ITP.
Collapse
Affiliation(s)
- Tzeon-Jye Chiou
- Department of Hematology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Fang Chang
- Department of Hematology, GCRC Laboratory, Mackay Memorial Hospital, Taipei, Taiwan
| | | | | | - Hsuan-Yu Lin
- National Cheng-Kung University Hospital, Tainan, Taiwan
| | | | | | | | - Yung-Chuan Sung
- Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung, Taiwan
| | | | - Li-Yuan Bai
- Institute Molecular Medicine, National Tsing-Hua University, Hsinchu, Taiwan
| | - Caleb G Chen
- Department of Hematology, Mackay Medical College, New Taipei, Taiwan.
| |
Collapse
|
170
|
Guglielmini G, Appolloni V, Momi S, De Groot PG, Battiston M, De Marco L, Falcinelli E, Gresele P. Matrix metalloproteinase-2 enhances platelet deposition on collagen under flow conditions. Thromb Haemost 2015; 115:333-43. [PMID: 26510894 DOI: 10.1160/th15-04-0300] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 09/18/2015] [Indexed: 11/05/2022]
Abstract
Platelets contain and release matrix metalloproteinase-2 (MMP-2) that in turn potentiates platelet aggregation. Platelet deposition on a damaged vascular wall is the first, crucial, step leading to thrombosis. Little is known about the effects of MMP-2 on platelet activation and adhesion under flow conditions. We studied the effect of MMP-2 on shear-dependent platelet activation using the O'Brien filtration system, and on platelet deposition using a parallel-plate perfusion chamber. Preincubation of human whole blood with active MMP-2 (50 ng/ml, i.e. 0.78 nM) shortened filter closure time (from 51.8 ± 3.6 sec to 40 ± 2.7 sec, p<0.05) and increased retained platelets (from 72.3 ± 2.3% to 81.1 ± 1.8%, p<0.05) in the O'Brien system, an effect prevented by a specific MMP-2 inhibitor. High shear stress induced the release of MMP-2 from platelets, while TIMP-2 levels were not significantly reduced, therefore, the MMP-2/TIMP-2 ratio increased significantly showing enhanced MMP-2 activity. Preincubation of whole blood with active MMP-2 (0.5 to 50 ng/ml, i.e 0.0078 to 0.78 nM) increased dose-dependently human platelet deposition on collagen under high shear-rate flow conditions (3000 sec⁻¹) (maximum +47.0 ± 11.9%, p<0.05, with 50 ng/ml), while pre-incubation with a MMP-2 inhibitor reduced platelet deposition. In real-time microscopy studies, increased deposition of platelets on collagen induced by MMP-2 started 85 sec from the beginning of perfusion, and was abolished by a GPIIb/IIIa antagonist, while MMP-2 had no effect on platelet deposition on fibrinogen or VWF. Confocal microscopy showed that MMP-2 enhances thrombus volume (+20.0 ± 3.0% vs control) rather than adhesion. In conclusion, we show that MMP-2 potentiates shear-induced platelet activation by enhancing thrombus formation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Paolo Gresele
- Paolo Gresele, MD, PhD, Section of Internal and Cardiovascular Medicine, Department of Medicine, University of Perugia, Via E. Dal Pozzo, 06126 Perugia, Italy, Tel.: +39 075 5783989, Fax: +39 075 5716083, E-mail:
| |
Collapse
|
171
|
Platelets induce apoptosis via membrane-bound FasL. Blood 2015; 126:1483-93. [PMID: 26232171 DOI: 10.1182/blood-2013-12-544445] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 07/06/2015] [Indexed: 02/06/2023] Open
Abstract
After tissue injury, both wound sealing and apoptosis contribute to restoration of tissue integrity and functionality. Although the role of platelets (PLTs) for wound closure and induction of regenerative processes is well established, the knowledge about their contribution to apoptosis is incomplete. Here, we show that PLTs present the death receptor Fas ligand (FasL) on their surface after activation. Activated PLTs as well as the isolated membrane fraction of activated PLTs but not of resting PLTs induced apoptosis in a dose-dependent manner in primary murine neuronal cells, human neuroblastoma cells, and mouse embryonic fibroblasts. Membrane protein from PLTs lacking membrane-bound FasL (FasL(△m/△m)) failed to induce apoptosis. Bax/Bak-mediated mitochondrial apoptosis signaling in target cells was not required for PLT-induced cell death, but increased the apoptotic response to PLT-induced Fas signaling. In vivo, PLT depletion significantly reduced apoptosis in a stroke model and an inflammation-independent model of N-methyl-d-aspartic acid-induced retinal apoptosis. Furthermore, experiments using PLT-specific PF4Cre(+) FasL(fl/fl) mice demonstrated a role of PLT-derived FasL for tissue apoptosis. Because apoptosis secondary to injury prevents inflammation, our findings describe a novel mechanism on how PLTs contribute to tissue homeostasis.
Collapse
|
172
|
Popp M, Thielmann I, Nieswandt B, Stegner D. Normal Platelet Integrin Function in Mice Lacking Hydrogen Peroxide-Induced Clone-5 (Hic-5). PLoS One 2015; 10:e0133429. [PMID: 26172113 PMCID: PMC4501753 DOI: 10.1371/journal.pone.0133429] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 06/26/2015] [Indexed: 11/21/2022] Open
Abstract
Integrin αIIbβ3 plays a central role in the adhesion and aggregation of platelets and thus is essential for hemostasis and thrombosis. Integrin activation requires the transmission of a signal from the small cytoplasmic tails of the α or β subunit to the large extracellular domains resulting in conformational changes of the extracellular domains to enable ligand binding. Hydrogen peroxide-inducible clone-5 (Hic-5), a member of the paxillin family, serves as a focal adhesion adaptor protein associated with αIIbβ3 at its cytoplasmic tails. Previous studies suggested Hic-5 as a novel regulator of integrin αIIbβ3 activation and platelet aggregation in mice. To assess this in more detail, we generated Hic-5-null mice and analyzed activation and aggregation of their platelets in vitro and in vivo. Surprisingly, lack of Hic-5 had no detectable effect on platelet integrin activation and function in vitro and in vivo under all tested conditions. These results indicate that Hic-5 is dispensable for integrin αIIbβ3 activation and consequently for arterial thrombosis and hemostasis in mice.
Collapse
Affiliation(s)
- Michael Popp
- Department of Experimental Biomedicine-Vascular Medicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Ina Thielmann
- Department of Experimental Biomedicine-Vascular Medicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Bernhard Nieswandt
- Department of Experimental Biomedicine-Vascular Medicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - David Stegner
- Department of Experimental Biomedicine-Vascular Medicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| |
Collapse
|
173
|
Mastenbroek TG, van Geffen JP, Heemskerk JWM, Cosemans JMEM. Acute and persistent platelet and coagulant activities in atherothrombosis. J Thromb Haemost 2015; 13 Suppl 1:S272-80. [PMID: 26149036 DOI: 10.1111/jth.12972] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The potential relevance of murine atherothrombosis models for understanding human disease has been debated in the past. Despite this, in the last decade, many thrombosis studies with atherogenic Apoe(-/-) mice have been performed, which provide novel insight into the molecular mechanisms by which platelet and coagulation processes accomplish acute thrombus formation after plaque disruption in vivo. Support for these mechanisms has come from whole blood flow perfusion studies over plaque material in vitro, which are also reviewed in this study. The main plaque-derived triggers for thrombus formation appear to be collagen and tissue factor, next to bioactive mediators such as prostaglandin E2. The atherothrombotic process relies on collagen- and ADP-receptor-induced platelet activation as well as on thrombin/fibrin generation via the extrinsic and intrinsic coagulation pathways. Less is known of the persistent effects of a thrombus on atherosclerosis progression, but evidence suggests roles herein of activated platelets and ongoing thrombin generation.
Collapse
Affiliation(s)
- T G Mastenbroek
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - J P van Geffen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - J W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - J M E M Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
174
|
Genome-wide association study of platelet aggregation in African Americans. BMC Genet 2015; 16:58. [PMID: 26024889 PMCID: PMC4448541 DOI: 10.1186/s12863-015-0217-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 05/13/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND We have previously shown that platelet aggregation has higher heritability in African Americans than European Americans. However, a genome-wide association study (GWAS) of platelet aggregation in African Americans has not been reported. We measured platelet aggregation in response to arachidonic acid, ADP, collagen, or epinephrine by optical aggregometry. The discovery cohort was 825 African Americans from the GeneSTAR study. Two replication cohorts were used: 119 African Americans from the Platelet Genes and Physiology Study and 1221 European Americans from GeneSTAR. Genotyping was conducted with Illumina 1 M arrays. For each cohort, age- and sex-adjusted linear mixed models were used to test for association between each SNP and each phenotype under an additive model. RESULTS Six SNPs were significantly associated with platelet aggregation (P<5×10(-8)) in the discovery sample. Of these, three SNPs in three different loci were confirmed: 1) rs12041331, in PEAR1 (platelet endothelial aggregation receptor 1), replicated in both African and European Americans for collagen- and epinephrine-induced aggregation, and in European Americans for ADP-induced aggregation; 2) rs11202221, in BMPR1A (bone morphogenetic protein receptor type1A), replicated in African Americans for ADP-induced aggregation; and 3) rs6566765 replicated in European Americans for ADP-induced aggregation. The rs11202221 and rs6566765 associations with agonist-induced platelet aggregation are novel. CONCLUSIONS In this first GWAS of agonist-induced platelet aggregation in African Americans, we discovered and replicated, novel associations of two variants with ADP-induced aggregation, and confirmed the association of a PEAR1 variant with multi-agonist-induced aggregation. Further study of these genes may provide novel insights into platelet biology.
Collapse
|
175
|
Platelet glycoprotein VI binds to polymerized fibrin and promotes thrombin generation. Blood 2015; 126:683-91. [PMID: 25977585 DOI: 10.1182/blood-2015-02-629717] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/27/2015] [Indexed: 11/20/2022] Open
Abstract
Fibrin, the coagulation end product, consolidates the platelet plug at sites of vascular injury and supports the recruitment of circulating platelets. In addition to integrin αIIbβ3, another as-yet-unidentified receptor is thought to mediate platelet interaction with fibrin. Platelet glycoprotein VI (GPVI) interacts with collagen and several other adhesive macromolecules. We evaluated the hypothesis that GPVI could be a functional platelet receptor for fibrin. Calibrated thrombin assays using platelet-rich plasma (PRP) showed that tissue factor-triggered thrombin generation was impaired in GPVI-deficient patients and reduced by the anti-GPVI Fab 9O12. Assays on reconstituted PRP and PRP from fibrinogen-deficient patients revealed a fibrinogen-dependent enhancement of thrombin generation, which relied on functional GPVI. The effect of GPVI was found to depend on fibrin polymerization. A binding assay showed a specific interaction between GPVI-Fc and fibrin, inhibited by the Fab 9O12. This Fab also reduced platelet adhesion to fibrin at low (300 s(-1)) and high (1500 s(-1)) wall shear rates. Platelets adherent to fibrin displayed shape change, exposure of procoagulant phospholipids, and the formation of small clots. When hirudinated blood was perfused at 1500 s(-1) over preformed fibrin-rich clots, the Fab 9O12 decreased the recruitment of platelets by up to 85%. This study identifies GPVI as a platelet receptor for polymerized fibrin with 2 major functions: (1) amplification of thrombin generation and (2) recruitment of circulating platelets to clots. These so-far-unrecognized properties of GPVI confer on it a key role in thrombus growth and stabilization.
Collapse
|
176
|
Platelet Gi protein Gαi2 is an essential mediator of thrombo-inflammatory organ damage in mice. Proc Natl Acad Sci U S A 2015; 112:6491-6. [PMID: 25944935 DOI: 10.1073/pnas.1505887112] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Platelets are crucial for hemostasis and thrombosis and exacerbate tissue injury following ischemia and reperfusion. Important regulators of platelet function are G proteins controlled by seven transmembrane receptors. The Gi protein Gα(i2) mediates platelet activation in vitro, but its in vivo role in hemostasis, arterial thrombosis, and postischemic infarct progression remains to be determined. Here we show that mice lacking Gα(i2) exhibit prolonged tail-bleeding times and markedly impaired thrombus formation and stability in different models of arterial thrombosis. We thus generated mice selectively lacking Gα(i2) in megakaryocytes and platelets (Gna(i2)(fl/fl)/PF4-Cre mice) and found bleeding defects comparable to those in global Gα(i2)-deficient mice. To examine the impact of platelet Gα(i2) in postischemic thrombo-inflammatory infarct progression, Gna(i2)(fl/fl)/PF4-Cre mice were subjected to experimental models of cerebral and myocardial ischemia/reperfusion injury. In the model of transient middle cerebral artery occlusion stroke Gna(i2)(fl/fl)/PF4-Cre mice developed significantly smaller brain infarcts and fewer neurological deficits than littermate controls. Following myocardial ischemia, Gna(i2)(fl/fl)/PF4-Cre mice showed dramatically reduced reperfusion injury which correlated with diminished formation of the ADP-dependent platelet neutrophil complex. In conclusion, our data provide definitive evidence that platelet Gα(i2) not only controls hemostatic and thrombotic responses but also is critical for the development of ischemia/reperfusion injury in vivo.
Collapse
|
177
|
Dütting S, Heidenreich J, Cherpokova D, Amin E, Zhang SC, Ahmadian MR, Brakebusch C, Nieswandt B. Critical off-target effects of the widely used Rac1 inhibitors NSC23766 and EHT1864 in mouse platelets. J Thromb Haemost 2015; 13:827-38. [PMID: 25628054 DOI: 10.1111/jth.12861] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 01/10/2015] [Indexed: 12/25/2022]
Abstract
BACKGROUND Platelet aggregation at sites of vascular injury is essential for normal hemostasis, but may also cause pathologic vessel occlusion. Rho GTPases are molecular switches that regulate essential cellular processes, and they have pivotal functions in the cardiovascular system. Rac1 is an important regulator of platelet cytoskeletal reorganization, and contributes to platelet activation. Rac1 inhibitors are thought to be beneficial in a wide range of therapeutic settings, and have therefore been tested in vivo for a variety of disorders. Two small-molecule inhibitors, NSC23766 and EHT1864, have been characterized in different cell types, demonstrating high specificity for Rac1 and Rac, respectively. OBJECTIVES To analyze the specificity of NSC23766 and EHT1864. METHODS Platelet function was assessed in mouse wild-type and Rac1-deficient platelets by the use of flow cytometric analysis of cellular activation and aggregometry. Platelet spreading was analyzed with differential interference contrast microscopy, and activation of effector molecules was analyzed with biochemical approaches. RESULTS NSC23766 and EHT1864 showed strong and distinct Rac1-independent effects at 100 μm in platelet function tests. Both inhibitors induced Rac1-specific inhibition of platelet spreading, but also markedly impaired agonist-induced activation of Rac1(-/-) platelets. Furthermore, glycoprotein Ib-mediated signaling was dramatically inhibited by NSC23766 in both wild-type and Rac1-deficient platelets. Importantly, these inhibitors directly affected the activation of the Rac1 effectors p21-activated kinase (PAK)1 and PAK2. CONCLUSIONS Our results reveal critical off-target effects of NSC23766 and EHT1864 at 100 μm in mammalian cells, raising questions about their utility as specific Rac1/Rac inhibitors in biochemical studies at these concentrations and possibly as therapeutic agents.
Collapse
Affiliation(s)
- S Dütting
- Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
178
|
Amantea D, Micieli G, Tassorelli C, Cuartero MI, Ballesteros I, Certo M, Moro MA, Lizasoain I, Bagetta G. Rational modulation of the innate immune system for neuroprotection in ischemic stroke. Front Neurosci 2015; 9:147. [PMID: 25972779 PMCID: PMC4413676 DOI: 10.3389/fnins.2015.00147] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 04/09/2015] [Indexed: 01/08/2023] Open
Abstract
The innate immune system plays a dualistic role in the evolution of ischemic brain damage and has also been implicated in ischemic tolerance produced by different conditioning stimuli. Early after ischemia, perivascular astrocytes release cytokines and activate metalloproteases (MMPs) that contribute to blood–brain barrier (BBB) disruption and vasogenic oedema; whereas at later stages, they provide extracellular glutamate uptake, BBB regeneration and neurotrophic factors release. Similarly, early activation of microglia contributes to ischemic brain injury via the production of inflammatory cytokines, including tumor necrosis factor (TNF) and interleukin (IL)-1, reactive oxygen and nitrogen species and proteases. Nevertheless, microglia also contributes to the resolution of inflammation, by releasing IL-10 and tumor growth factor (TGF)-β, and to the late reparative processes by phagocytic activity and growth factors production. Indeed, after ischemia, microglia/macrophages differentiate toward several phenotypes: the M1 pro-inflammatory phenotype is classically activated via toll-like receptors or interferon-γ, whereas M2 phenotypes are alternatively activated by regulatory mediators, such as ILs 4, 10, 13, or TGF-β. Thus, immune cells exert a dualistic role on the evolution of ischemic brain damage, since the classic phenotypes promote injury, whereas alternatively activated M2 macrophages or N2 neutrophils prompt tissue remodeling and repair. Moreover, a subdued activation of the immune system has been involved in ischemic tolerance, since different preconditioning stimuli act via modulation of inflammatory mediators, including toll-like receptors and cytokine signaling pathways. This further underscores that the immuno-modulatory approach for the treatment of ischemic stroke should be aimed at blocking the detrimental effects, while promoting the beneficial responses of the immune reaction.
Collapse
Affiliation(s)
- Diana Amantea
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria Rende, Italy
| | | | - Cristina Tassorelli
- C. Mondino National Neurological Institute Pavia, Italy ; Department of Brain and Behavioral Sciences, University of Pavia Pavia, Italy
| | - María I Cuartero
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid and Instituto de Investigación Hospital 12 de Octubre Madrid, Spain
| | - Iván Ballesteros
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid and Instituto de Investigación Hospital 12 de Octubre Madrid, Spain
| | - Michelangelo Certo
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria Rende, Italy
| | - María A Moro
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid and Instituto de Investigación Hospital 12 de Octubre Madrid, Spain
| | - Ignacio Lizasoain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid and Instituto de Investigación Hospital 12 de Octubre Madrid, Spain
| | - Giacinto Bagetta
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria Rende, Italy ; Section of Neuropharmacology of Normal and Pathological Neuronal Plasticity, University Consortium for Adaptive Disorders and Head Pain, University of Calabria Rende, Italy
| |
Collapse
|
179
|
Qu L, Jiang M, Qiu W, Lu S, Zhao Y, Xia L, Ruan C, Zhao Y. Assessment of the Diagnostic Value of Plasma Levels, Activities, and Their Ratios of von Willebrand Factor and ADAMTS13 in Patients with Cerebral Infarction. Clin Appl Thromb Hemost 2015; 22:252-9. [PMID: 25916953 DOI: 10.1177/1076029615583347] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Raised levels of von Willebrand factor (VWF) and reduced levels of a disintegrin and a metalloproteinase with a thrombospondin type I motif, member 13 (ADAMTS13) activity are associated with thrombosis. We aimed to investigate the relationships between plasma levels of VWF and ADAMTS13, their ratios, and the occurrence of cerebral infarction and to understand the roles of VWF and ADAMTS13 in cerebral infarction. METHODS Ninety-four patients with cerebral infarction and 103 controls were analyzed. Plasma levels of VWF antigen (VWF: Ag), VWF ristocetin cofactor activity (VWF: Rcof), and VWF collagen binding activity (VWF: CB) were measured by enzyme-linked immunosorbent assay (ELISA). The ADAMTS13 activity (ADAMTS13) was measured with FRETS-VWF73. The relationship between plasma levels and ratios of VWF and ADAMTS13 and the occurrence of cerebral infarction were analyzed. RESULTS Patients with cerebral infarction displayed higher VWF: Ag and VWF: Rcof levels and lower ADAMTS13, VWF: CB/VWF: Ag, ADAMTS13/VWF: Ag, and ADAMTS13/VWF: Rcof levels compared to controls (P < .01). The highest quartiles of VWF: Ag (odds ratio [OR] = 5.11, 95% confidence interval [CI], 1.49-17.50) and VWF: Rcof (OR = 5.04, 95% CI, 1.62-15.66) and the lowest quartiles of VWF: CB/VWF: Ag (OR = 5.91, 95% CI, 1.95-17.93), ADAMTS13/VWF: Ag (OR = 9.11, 95% CI, 2.49-33.33), and ADAMTS13/VWF: Rcof (OR = 3.73, 95% CI, 1.39-10.03) are associated with cerebral infarction. CONCLUSIONS An association was found between reduced levels of VWF: CB/VWF: Ag, ADAMTS13/VWF: Ag, and ADAMTS13/VWF: Rcof ratios and cerebral infarction. Our data suggest that increased levels of VWF and reduced levels of ADAMTS13 activity may contribute to the pathogenesis of cerebral infarction.
Collapse
Affiliation(s)
- Le Qu
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Suzhou, China
| | - Miao Jiang
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Suzhou, China
| | - Wenjuan Qiu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shiqi Lu
- Department of Emergency, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yunxiao Zhao
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Suzhou, China
| | - Lijun Xia
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Suzhou, China
| | - Changgeng Ruan
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Suzhou, China
| | - Yiming Zhao
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Suzhou, China
| |
Collapse
|
180
|
Targeted downregulation of platelet CLEC-2 occurs through Syk-independent internalization. Blood 2015; 125:4069-77. [PMID: 25795918 DOI: 10.1182/blood-2014-11-611905] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 03/13/2015] [Indexed: 12/16/2022] Open
Abstract
Platelet aggregation at sites of vascular injury is not only essential for hemostasis, but may also cause acute ischemic disease states such as myocardial infarction or stroke. The hemi-immunoreceptor tyrosine-based activation motif-containing C-type lectinlike receptor 2 (CLEC-2) mediates powerful platelet activation through a Src- and spleen tyrosine kinase (Syk)-dependent tyrosine phosphorylation cascade. Thereby, CLEC-2 not only contributes to thrombus formation and stabilization but also plays a central role in blood-lymphatic vessel development, tumor metastasis, and prevention of inflammatory bleeding, making it a potential pharmacologic target to modulate these processes. We have previously shown that injection of the anti-CLEC-2 antibody, INU1, results in virtually complete immunodepletion of platelet CLEC-2 in mice, which is, however, preceded by a severe transient thrombocytopenia thereby limiting its potential therapeutic use. The mechanisms underlying this targeted CLEC-2 downregulation have remained elusive. Here, we show that INU1-induced CLEC-2 immunodepletion occurs through Src-family kinase-dependent receptor internalization in vitro and in vivo, presumably followed by intracellular degradation. In mice with platelet-specific Syk deficiency, INU1-induced CLEC-2 internalization/degradation was fully preserved whereas the associated thrombocytopenia was largely prevented. These results show for the first time that CLEC-2 can be downregulated from the platelet surface through internalization in vitro and in vivo and that this can be mechanistically uncoupled from the associated antibody-induced thrombocytopenia.
Collapse
|
181
|
Nording HM, Seizer P, Langer HF. Platelets in inflammation and atherogenesis. Front Immunol 2015; 6:98. [PMID: 25798138 PMCID: PMC4351644 DOI: 10.3389/fimmu.2015.00098] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/19/2015] [Indexed: 12/12/2022] Open
Abstract
Platelets contribute to processes beyond thrombus formation and may play a so far underestimated role as an immune cell in various circumstances. This review outlines immune functions of platelets in host defense, but also how they may contribute to mechanisms of infectious diseases. A particular emphasis is placed on the interaction of platelets with other immune cells. Furthermore, this article outlines the features of atherosclerosis as an inflammatory vascular disease highlighting the role of platelet crosstalk with cellular and soluble factors involved in atheroprogression. Understanding, how platelets influence these processes of vascular remodeling will shed light on their role for tissue homeostasis beyond intravascular thrombosis. Finally, translational implications of platelet-mediated inflammation in atherosclerosis are discussed.
Collapse
Affiliation(s)
- Henry M. Nording
- University Clinic for Cardiology and Cardiovascular Medicine, Eberhard Karls-University Tübingen, Tübingen, Germany
- Section for Cardioimmunology, Eberhard Karls-University Tübingen, Tübingen, Germany
| | - Peter Seizer
- University Clinic for Cardiology and Cardiovascular Medicine, Eberhard Karls-University Tübingen, Tübingen, Germany
| | - Harald F. Langer
- University Clinic for Cardiology and Cardiovascular Medicine, Eberhard Karls-University Tübingen, Tübingen, Germany
- Section for Cardioimmunology, Eberhard Karls-University Tübingen, Tübingen, Germany
| |
Collapse
|
182
|
Platelet transfusions in platelet consumptive disorders are associated with arterial thrombosis and in-hospital mortality. Blood 2015; 125:1470-6. [PMID: 25588677 DOI: 10.1182/blood-2014-10-605493] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
While platelets are primary mediators of hemostasis, there is emerging evidence to show that they may also mediate pathologic thrombogenesis. Little data are available on risks and benefits associated with platelet transfusions in thrombotic thrombocytopenic purpura (TTP), heparin-induced thrombocytopenia (HIT) and immune thrombocytopenic purpura (ITP). This study utilized the Nationwide Inpatient Sample to evaluate the current in-hospital platelet transfusion practices and their association with arterial/venous thrombosis, acute myocardial infarction (AMI), stroke, and in-hospital mortality over 5 years (2007-2011). Age and gender-adjusted odds ratios (adjOR) associated with platelet transfusions were calculated. There were 10 624 hospitalizations with TTP; 6332 with HIT and 79 980 with ITP. Platelet transfusions were reported in 10.1% TTP, 7.1% HIT, and 25.8% ITP admissions. Platelet transfusions in TTP were associated with higher odds of arterial thrombosis (adjOR = 5.8, 95%CI = 1.3-26.6), AMI (adjOR = 2.0, 95%CI = 1.2-3.3) and mortality (adjOR = 2.0,95%CI = 1.3-3.0), but not venous thrombosis. Platelet transfusions in HIT were associated with higher odds of arterial thrombosis (adjOR = 3.4, 95%CI = 1.2-9.5) and mortality (adjOR = 5.2, 95%CI = 2.6-10.5) but not venous thrombosis. Except for AMI, all relationships remained significant after adjusting for clinical severity and acuity. No associations were significant for ITP. Platelet transfusions are associated with higher odds of arterial thrombosis and mortality among TTP and HIT patients.
Collapse
|
183
|
Xu X. Plant Polysaccharides and Their Effects on Cell Adhesion. POLYSACCHARIDES 2015. [DOI: 10.1007/978-3-319-16298-0_67] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
184
|
Liao CY, Lee CL, Wang HC, Liang SS, Kung PH, Wu YC, Chang FR, Wu CC. CLL2-1, a chemical derivative of orchid 1,4-phenanthrenequinones, inhibits human platelet aggregation through thiol modification of calcium-diacylglycerol guanine nucleotide exchange factor-I (CalDAG-GEFI). Free Radic Biol Med 2015; 78:101-10. [PMID: 25451646 DOI: 10.1016/j.freeradbiomed.2014.10.512] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 10/14/2014] [Accepted: 10/14/2014] [Indexed: 11/29/2022]
Abstract
CalDAG-GEFI is a guanine nucleotide exchange factor, which actives small GTPase Rap1 and plays an important role in platelet aggregation. Our previous study has shown that CalDAG-GEFI contains redox-sensitive thiols, and its function can be inhibited by thiol modification. In the present study, the effect of CLL2-1, a 1,4-phenanthrenequinone, on CalDAG-GEFI and platelet functions was investigated. In human platelets, CLL2-1 prevented platelet aggregation caused by various stimulators. Flow cytometric analysis revealed that CLL2-1 inhibited GPIIb/IIIa activation and P-selectin secretion. Moreover, CLL2-1 prevented Rap1 activation caused by thrombin, the Ca(2+) ionophore A23187, and the diacylglycerol mimetic phorbol 12-myristate 13-acetate, while only slightly inhibited thrombin-induced increases in [Ca(2+)]i and did not inhibit protein kinase C activation. Western blots after reducing SDS-PAGE showed that treatment of either platelets or platelet lysates with CLL2-1 led to a decrease of monomeric CalDAG-GEFI and appearance of cross-linked oligomers of CalDAG-GEFI, and these effects were inhibited by pretreatment of platelets or lysates with thiol reducing agents prior to the addition of CLL2-1, indicating thiol modification of CalDAG-GEFI by CLL2-1. Furthermore, the thiol reducing agents also prevented the inhibitory effect of CLL2-1 on Rap1 activation, GPIIb/IIIa activation, and platelet aggregation. In CalDAG-GEFI-overexpressing human embryonic kidney 293T cells, CLL2-1 also inhibited CalDAG-GEFI-mediated Rap1 activation. Taken together, our results suggest that the antiplatelet effect of CLL2-1 is due to, at least in part, inhibition of CalDAG-GEFI-mediated Rap1 activation, and provide the basis for development of novel antiplatelet drugs.
Collapse
Affiliation(s)
- Chieh-Yu Liao
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Lin Lee
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan
| | - Hui-Chun Wang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shih-Shin Liang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Hsiung Kung
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yang-Chang Wu
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chin-Chung Wu
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan.
| |
Collapse
|
185
|
Srihirun S, Tanjararak N, Chuncharunee S, Sritara P, Kaewvichit R, Fucharoen S, Pattanapanyasat K, Sibmooh N. Platelet hyperactivity in thalassemia patients with elevated tricuspid regurgitant velocity and the association with hemolysis. Thromb Res 2015; 135:121-6. [DOI: 10.1016/j.thromres.2014.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/06/2014] [Accepted: 10/08/2014] [Indexed: 01/19/2023]
|
186
|
Hofmann S, Braun A, Pozgaj R, Morowski M, Vögtle T, Nieswandt B. Mice lacking the SLAM family member CD84 display unaltered platelet function in hemostasis and thrombosis. PLoS One 2014; 9:e115306. [PMID: 25551754 PMCID: PMC4281120 DOI: 10.1371/journal.pone.0115306] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 11/23/2014] [Indexed: 01/25/2023] Open
Abstract
Background Platelets are anuclear cell fragments derived from bone marrow megakaryocytes that safeguard vascular integrity by forming thrombi at sites of vascular injury. Although the early events of thrombus formation—platelet adhesion and aggregation—have been intensively studied, less is known about the mechanisms and receptors that stabilize platelet-platelet interactions once a thrombus has formed. One receptor that has been implicated in this process is the signaling lymphocyte activation molecule (SLAM) family member CD84, which can undergo homophilic interactions and becomes phosphorylated upon platelet aggregation. Objective The role of CD84 in platelet physiology and thrombus formation was investigated in CD84-deficient mice. Methods and Results We generated CD84-deficient mice and analyzed their platelets in vitro and in vivo. Cd84−/− platelets exhibited normal activation and aggregation responses to classical platelet agonists. Furthermore, CD84 deficiency did not affect integrin-mediated clot retraction and spreading of activated platelets on fibrinogen. Notably, also the formation of stable three-dimensional thrombi on collagen-coated surfaces under flow ex vivo was unaltered in the blood of Cd84−/− mice. In vivo, Cd84−/− mice exhibited unaltered hemostatic function and arterial thrombus formation. Conclusion These results show that CD84 is dispensable for thrombus formation and stabilization, indicating that its deficiency may be functionally compensated by other receptors or that it may be important for platelet functions different from platelet-platelet interactions.
Collapse
Affiliation(s)
- Sebastian Hofmann
- University of Würzburg, Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| | - Attila Braun
- University of Würzburg, Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| | - Rastislav Pozgaj
- University of Würzburg, Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| | - Martina Morowski
- University of Würzburg, Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, Würzburg, Germany
| | - Timo Vögtle
- University of Würzburg, Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, Würzburg, Germany
- * E-mail: (BN); (TV)
| | - Bernhard Nieswandt
- University of Würzburg, Department of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, Würzburg, Germany
- * E-mail: (BN); (TV)
| |
Collapse
|
187
|
Abstract
Thrombotic and cardiovascular events are among the leading causes of death for patients with polycythemia vera (PV), and thrombosis history is a key criterion for patient risk stratification and treatment strategy. Little is known, however, about mechanisms of thrombogenesis in patients with PV. This report provides an overview of thrombogenesis pathophysiology in patients with PV and elucidates the roles of conventional and nonconventional thrombotic risk factors. In addition to several conventional risk factors for thrombosis, clinical data have implicated increased hematocrit and red blood cell adhesiveness, activated platelets, leukocytosis, and elevated JAK2(V617F) allele burden in patients with PV. Furthermore, PV-related inflammation may exacerbate thrombogenesis through varied mechanisms, including endothelial damage, inhibition of natural anticoagulant pathways, and secretion of procoagulant factors. These findings suggest a direct link between myeloproliferation and thrombogenesis in PV, which is likely to provide new opportunities for targeted antithrombotic interventions aimed at decreasing PV-related morbidity and mortality.
Collapse
|
188
|
A novel role of Eruca sativa Mill. (rocket) extract: antiplatelet (NF-κB inhibition) and antithrombotic activities. Nutrients 2014; 6:5839-52. [PMID: 25514563 PMCID: PMC4277002 DOI: 10.3390/nu6125839] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 11/24/2014] [Accepted: 11/25/2014] [Indexed: 11/17/2022] Open
Abstract
Background: Epidemiological studies have shown the prevention of cardiovascular diseases through the regular consumption of vegetables. Eruca sativa Mill., commonly known as rocket, is a leafy vegetable that has anti-inflammatory activity. However, its antiplatelet and antithrombotic activities have not been described. Methods: Eruca sativa Mill. aqueous extract (0.1 to 1 mg/mL), was evaluated on human platelets: (i) P-selectin expression by flow cytometry; (ii) platelet aggregation induced by ADP, collagen and arachidonic acid; (iii) IL-1β, TGF-β1, CCL5 and thromboxane B2 release; and (iv) activation of NF-κB and PKA by western blot. Furthermore, (v) antithrombotic activity (200 mg/kg) and (vi) bleeding time in murine models were evaluated. Results: Eruca sativa Mill. aqueous extract (0.1 to 1 mg/mL) inhibited P-selectin expression and platelet aggregation induced by ADP. The release of platelet inflammatory mediators (IL-1β, TGF-β1, CCL5 and thromboxane B2) induced by ADP was inhibited by Eruca sativa Mill. aqueous extract. Furthermore, Eruca sativa Mill. aqueous extract inhibited NF-κB activation. Finally, in murine models, Eruca sativa Mill. aqueous extract showed significant antithrombotic activity and a slight effect on bleeding time. Conclusion: Eruca sativa Mill. presents antiplatelet and antithrombotic activity.
Collapse
|
189
|
Shovlin CL. Pulmonary arteriovenous malformations. Am J Respir Crit Care Med 2014; 190:1217-28. [PMID: 25420112 PMCID: PMC4315816 DOI: 10.1164/rccm.201407-1254ci] [Citation(s) in RCA: 162] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 10/20/2014] [Indexed: 11/16/2022] Open
Abstract
Within the past decade, pulmonary arteriovenous malformations (PAVMs) have evolved from rare curiosities to not uncommon clinical states, with the latest estimates suggesting a prevalence of ~1 in 2,600. PAVMs provide anatomic right-to-left shunts, allowing systemic venous blood to bypass gas exchange and pulmonary capillary bed processing. Hypoxemia and enhanced ventilatory demands result, although both are usually asymptomatic. Paradoxical emboli lead to strokes and cerebral abscesses, and these commonly occur in individuals with previously undiagnosed PAVMs. PAVM hemorrhage is rare but is the main cause of maternal death in pregnancy. PAVM occlusion by embolization is the standard of care to reduce these risks. However, recent data demonstrate that currently recommended management protocols can result in levels of radiation exposure that would be classified as harmful. Recent publications also provide a better appreciation of the hematologic and cardiovascular demands required to maintain arterial oxygen content and oxygen consumption in hypoxemic patients, identify patient subgroups at higher risk of complications, and emphasize the proportion of radiologically visible PAVMs too small to treat by embolization. This review, therefore, outlines medical states that exacerbate the consequences of PAVMs. Chief among these is iron deficiency, which is commonly present due to concurrent hereditary hemorrhagic telangiectasia: iron deficiency impairs hypoxemia compensations by restricting erythropoiesis and increases the risk of ischemic strokes. Management of periodontal disease, dental interventions, pulmonary hypertension, and pregnancy also requires specific consideration in the setting of PAVMs. The review concludes by discussing to what extent previously recommended protocols may benefit from modification or revision.
Collapse
Affiliation(s)
- Claire L. Shovlin
- NHLI Cardiovascular Sciences, Imperial College, London; and
- Respiratory Medicine, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
190
|
Purinergic control of inflammation and thrombosis: Role of P2X1 receptors. Comput Struct Biotechnol J 2014; 13:106-10. [PMID: 25709760 PMCID: PMC4334884 DOI: 10.1016/j.csbj.2014.11.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/21/2014] [Accepted: 11/24/2014] [Indexed: 01/08/2023] Open
Abstract
Inflammation shifts the hemostatic mechanisms in favor of thrombosis. Upon tissue damage or infection, a sudden increase of extracellular ATP occurs, that might contribute to the crosstalk between inflammation and thrombosis. On platelets, P2X1 receptors act to amplify platelet activation and aggregation induced by other platelet agonists. These receptors critically contribute to thrombus stability in small arteries. Besides platelets, studies by our group indicate that these receptors are expressed by neutrophils. They promote neutrophil chemotaxis, both in vitro and in vivo. In a laser-induced injury mouse model of thrombosis, it appears that neutrophils are required to initiate thrombus formation and coagulation activation on inflamed arteriolar endothelia. In this model, by using P2X1−/ − mice, we recently showed that P2X1 receptors, expressed on platelets and neutrophils, play a key role in thrombus growth and fibrin generation. Intriguingly, in a model of endotoxemia, P2X1−/ − mice exhibited aggravated oxidative tissue damage, along with exacerbated thrombocytopenia and increased activation of coagulation, which translated into higher susceptibility to septic shock. Thus, besides its ability to recruit neutrophils and platelets on inflamed endothelia, the P2X1 receptor also contributes to limit the activation of circulating neutrophils under systemic inflammatory conditions. Taken together, these data suggest that P2X1 receptors are involved in the interplay between platelets, neutrophils and thrombosis. We propose that activation of these receptors by ATP on neutrophils and platelets represents a new mechanism that regulates thrombo-inflammation.
Collapse
|
191
|
Abstract
We aimed to elucidate platelet function in trauma patients, as it is pivotal for hemostasis yet remains scarcely investigated in this population. We conducted a prospective observational study of platelet aggregation capacity in 213 adult trauma patients on admission to an emergency department (ED). Inclusion criteria were trauma team activation and arterial cannula insertion on arrival. Blood samples were analyzed by multiple electrode aggregometry initiated by thrombin receptor agonist peptide 6 (TRAP) or collagen using a Multiplate device. Blood was sampled median 65 min after injury; median injury severity score (ISS) was 17; 14 (7%) patients received 10 or more units of red blood cells in the ED (massive transfusion); 24 (11%) patients died within 28 days of trauma: 17 due to cerebral injuries, four due to exsanguination, and three from other causes. No significant association was found between aggregation response and ISS. Higher TRAP values were associated with death due to cerebral injuries (P < 0.01, when corrected for ISS and platelet counts), whereas lower platelet counts were associated with massive transfusion (P < 0.01, when corrected for ISS and aggregation). An aggregation value of 145 IU by TRAP significantly identified death due to cerebral injury (sensitivity 71% and specificity 76%, P < 0.01) by receiver operating characteristic-curve analysis; the corresponding value of platelet counts for massive transfusion was 189 × 10/l (sensitivity 86%, specificity 75%, P < 0.01). We concluded there was no simple relationship between platelet aggregation and injury severity. Our results indicate that high platelet aggregation values are associated with fatality of cerebral injury.
Collapse
|
192
|
Schunke KJ, Toung TK, Zhang J, Pathak AP, Xu J, Zhang J, Koehler RC, Faraday N. A novel atherothrombotic model of ischemic stroke induced by injection of collagen into the cerebral vasculature. J Neurosci Methods 2014; 239:65-74. [PMID: 25314906 DOI: 10.1016/j.jneumeth.2014.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/02/2014] [Accepted: 10/03/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Most ischemic strokes in humans are caused by ruptured arterial atheroma, which activate platelets and produce thrombi that occlude cerebral vessels. METHODS To simulate these events, we threaded a catheter through the internal carotid artery toward the middle cerebral artery (MCA) orifice and injected collagen directly into the cerebral circulation of male C57Bl/6 mice and Wistar rats. RESULTS Laser-Doppler flowmetry demonstrated reductions in cerebral blood flow (CBF) of ∼80% in mice and ∼60% in rats. CBF spontaneously increased but remained depressed after catheter withdrawal. Magnetic resonance imaging showed that ipsilateral CBF was reduced at 3h after collagen injection and markedly improved at 48 h. Micro-computed tomography revealed reduced blood vessel density in the ipsilateral MCA territory at 3 h. Gross examination of excised brains revealed thrombi within ipsilateral cerebral arteries at 3 h, but not 24 h, after collagen injection. Immunofluorescence microscopy confirmed that platelets and fibrinogen/fibrin were major components of these thrombi at both macrovascular and microvascular levels. Cerebral infarcts comprising ∼30% of hemispheric volume and neurobehavioral deficits were observed 48 h after ischemic injury in both mice and rats. COMPARISON WITH EXISTING METHODS Collagen injection caused brain injury that was similar in magnitude and variability to mechanical MCA occlusion or injection of a pre-formed clot; however, alterations in CBF and the mechanism of vascular occlusion were more consistent with clinical ischemic stroke. CONCLUSION This novel rodent model of ischemic stroke has pathophysiologic characteristics consistent with clinical atherothrombotic stroke, is technically feasible, and creates reproducible brain injury.
Collapse
Affiliation(s)
- Kathryn J Schunke
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas K Toung
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jian Zhang
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Arvind P Pathak
- The Russell H. Morgan Department of Radiology & Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jiadi Xu
- F. M. Kirby Functional Imaging Center, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Jiangyang Zhang
- The Russell H. Morgan Department of Radiology & Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Raymond C Koehler
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nauder Faraday
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
193
|
SLAP/SLAP2 prevent excessive platelet (hem)ITAM signaling in thrombosis and ischemic stroke in mice. Blood 2014; 125:185-94. [PMID: 25301707 DOI: 10.1182/blood-2014-06-580597] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Glycoprotein VI and C-type lectin-like receptor 2 are essential platelet activating receptors in hemostasis and thrombo-inflammatory disease, which signal through a (hem)immunoreceptor tyrosine-based activation motif (ITAM)-dependent pathway. The adapter molecules Src-like adapter proteins (SLAP and SLAP2) are involved in the regulation of immune cell surface expression and signaling, but their function in platelets is unknown. In this study, we show that platelets expressed both SLAP isoforms and that overexpression of either protein in a heterologous cell line almost completely inhibited glycoprotein VI and C-type lectin-like receptor 2 signaling. In mice, single deficiency of SLAP or SLAP2 had only moderate effects on platelet function, whereas double deficiency of both adapters resulted in markedly increased signal transduction, integrin activation, granule release, aggregation, procoagulant activity, and thrombin generation in response to (hem)ITAM-coupled, but not G protein-coupled, receptor activation. In vivo, constitutive SLAP/SLAP2 knockout mice displayed accelerated occlusive arterial thrombus formation and a dramatically worsened outcome after focal cerebral ischemia. This was attributed to the absence of both adapter proteins in platelets, as demonstrated by adoptive transfer of Slap(-/-)/Slap2(-/-) platelets into wild-type mice. Our results establish SLAP and SLAP2 as critical inhibitors of platelet (hem)ITAM signaling in the setting of arterial thrombosis and ischemic stroke.
Collapse
|
194
|
The adaptor protein Swiprosin-1/EFhd2 is dispensable for platelet function in mice. PLoS One 2014; 9:e107139. [PMID: 25243606 PMCID: PMC4170979 DOI: 10.1371/journal.pone.0107139] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 08/06/2014] [Indexed: 11/19/2022] Open
Abstract
Background Platelets are anuclear cell fragments derived from bone marrow megakaryocytes that safeguard vascular integrity, but may also cause pathological vessel occlusion. Reorganizations of the platelet cytoskeleton and agonist-induced intracellular Ca2+-mobilization are crucial for platelet hemostatic function. EF-hand domain containing 2 (EFhd2, Swiprosin-1) is a Ca2+-binding cytoskeletal adaptor protein involved in actin remodeling in different cell types, but its function in platelets is unknown. Objective Based on the described functions of EFhd2 in immune cells, we tested the hypothesis that EFhd2 is a crucial adaptor protein for platelet function acting as a regulator of Ca2+-mobilization and cytoskeletal rearrangements. Methods and Results We generated EFhd2-deficient mice and analyzed their platelets in vitro and in vivo. Efhd2-/- mice displayed normal platelet count and size, exhibited an unaltered in vivo life span and showed normal Ca2+-mobilization and activation/aggregation responses to classic agonists. Interestingly, upon stimulation of the immunoreceptor tyrosine-based activation motif-coupled receptor glycoprotein (GP) VI, Efhd2-/- platelets showed a slightly increased coagulant activity. Furthermore, absence of EFhd2 had no significant impact on integrin-mediated clot retraction, actomyosin rearrangements and spreading of activated platelets on fibrinogen. In vivo EFhd2-deficiency resulted in unaltered hemostatic function and unaffected arterial thrombus formation. Conclusion These results show that EFhd2 is not essential for platelet function in mice indicating that other cytoskeletal adaptors may functionally compensate its loss.
Collapse
|
195
|
Protection of glycocalyx decreases platelet adhesion after ischaemia/reperfusion. Eur J Anaesthesiol 2014; 31:474-81. [DOI: 10.1097/eja.0000000000000085] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
196
|
Zhang L, Sun Y. Biomimetic design of platelet adhesion inhibitors to block integrin α2β1-collagen interactions: I. Construction of an affinity binding model. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2014; 30:4725-4733. [PMID: 24697616 DOI: 10.1021/la404599s] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Platelet adhesion on a collagen surface through integrin α2β1 has been proven to be significant for the formation of arterial thrombus. However, the molecular determinants mediating the integrin-collagen complex remain unclear. In the present study, the dynamics of integrin-collagen binding and molecular interactions were investigated using molecular dynamics (MD) simulations and molecular mechanics-Poisson-Boltzmann surface area (MM-PBSA) analysis. Hydrophobic interaction is identified as the major driving force for the formation of the integrin-collagen complex. On the basis of the MD simulation and MM-PBSA results, an affinity binding model (ABM) of integrin for collagen is constructed; it is composed of five residues, including Y157, N154, S155, R288, and L220. The ABM has been proven to capture the major binding motif contributing 84.8% of the total binding free energy. On the basis of the ABM, we expect to establish a biomimetic design strategy of platelet adhesion inhibitors, which would be beneficial for the development of potent peptide-based drugs for thrombotic diseases.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University , Tianjin 300072, People's Republic of China
| | | |
Collapse
|
197
|
Zhang L, Zhang C, Sun Y. Biomimetic design of platelet adhesion inhibitors to block integrin α2β1-collagen interactions: II. Inhibitor library, screening, and experimental validation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2014; 30:4734-4742. [PMID: 24697658 DOI: 10.1021/la4046012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Platelet adhesion on collagen mediated by integrin α2β1 has been proven important in arterial thrombus formation, leading to an exigent demand on development of potent inhibitors for the integrin α2β1-collagen binding. In the present study, a biomimetic design strategy of platelet adhesion inhibitors was established, based on the affinity binding model of integrin proposed in part I. First, a heptapeptide library containing 8000 candidates was designed to functionally mimic the binding motif of integrin α2β1. Then, each heptapeptide in the library was docked onto a collagen molecule for the assessment of its affinity, followed by a screening based on its structure similarity to the original structure in the affinity binding model. Eight candidates were then selected for further screening by molecular dynamics (MD) simulations. Thereafter, three candidates chosen from MD simulations were separately added into the physiological saline containing separated integrin and collagen, to check their abilities for blocking the integrin-collagen interaction using MD simulations. Of these three candidates, significant inhibition was observed in the presence of LWWNSYY. Finally, the binding affinity of LWWNSYY for collagen was demonstrated by isothermal titration calorimetry. Moreover, significant inhibition of platelet adhesion in the presence of LWWNSYY has been experimentally validated. This work has thus developed an effective strategy for the biomimetic design of peptide-based platelet adhesion inhibitors.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University , Tianjin 300072, People's Republic of China
| | | | | |
Collapse
|
198
|
Bender M, Falet H. Post-translational arginylation as a novel regulator of platelet function. Haematologica 2014; 99:402-4. [PMID: 24598850 DOI: 10.3324/haematol.2013.103119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
199
|
Gawaz M, Vogel S, Pfannenberg C, Pichler B, Langer H, Bigalke B. Implications of glycoprotein VI for theranostics. Thromb Haemost 2014; 112:26-31. [PMID: 24553806 DOI: 10.1160/th13-09-0756] [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: 09/13/2013] [Accepted: 01/31/2014] [Indexed: 01/03/2023]
Abstract
Glycoprotein VI (GPVI), a membrane glycoprotein solely expressed in platelets and megakaryocytes, plays a critical role in thrombus formation due to collagen/GPVI-mediated platelet activation and adhesion. Recent studies have shown that surface expression of GPVI on circulating platelets is enhanced in acute cardiovascular diseases such as myocardial infarction and ischaemic stroke. Increased GPVI levels are associated with poor clinical outcome and are an early indicator for imminent myocardial infarction in patients with chest pain. The soluble form of the dimeric GPVI fusion protein (sGPVI-Fc) binds with high affinity to collagen and atherosclerotic plaque tissue. Non-invasive imaging studies with radiolabelled sGPVI-Fc show specific binding activity to vascular lesions in vivo. Further, sGPVI-Fc has been developed as a new therapeutic platelet-based strategy for lesion-directed antithrombotic therapy. This review summarises the potential of GPVI for diagnostic and therapeutic options based on novel non-invasive molecular imaging modalities to ameliorate care of patients with cardiovascular diseases.
Collapse
Affiliation(s)
- M Gawaz
- Meinrad Gawaz, MD, Department of Cardiology, University of Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany, Tel.: +49 7071 29 83688, Fax: +49 7071 29 5749, E-mail:
| | | | | | | | | | | |
Collapse
|
200
|
Ischaemic strokes in patients with pulmonary arteriovenous malformations and hereditary hemorrhagic telangiectasia: associations with iron deficiency and platelets. PLoS One 2014; 9:e88812. [PMID: 24586400 PMCID: PMC3929507 DOI: 10.1371/journal.pone.0088812] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 01/13/2014] [Indexed: 01/24/2023] Open
Abstract
Background Pulmonary first pass filtration of particles marginally exceeding ∼7 µm (the size of a red blood cell) is used routinely in diagnostics, and allows cellular aggregates forming or entering the circulation in the preceding cardiac cycle to lodge safely in pulmonary capillaries/arterioles. Pulmonary arteriovenous malformations compromise capillary bed filtration, and are commonly associated with ischaemic stroke. Cohorts with CT-scan evident malformations associated with the highest contrast echocardiographic shunt grades are known to be at higher stroke risk. Our goal was to identify within this broad grouping, which patients were at higher risk of stroke. Methodology 497 consecutive patients with CT-proven pulmonary arteriovenous malformations due to hereditary haemorrhagic telangiectasia were studied. Relationships with radiologically-confirmed clinical ischaemic stroke were examined using logistic regression, receiver operating characteristic analyses, and platelet studies. Principal Findings Sixty-one individuals (12.3%) had acute, non-iatrogenic ischaemic clinical strokes at a median age of 52 (IQR 41–63) years. In crude and age-adjusted logistic regression, stroke risk was associated not with venous thromboemboli or conventional neurovascular risk factors, but with low serum iron (adjusted odds ratio 0.96 [95% confidence intervals 0.92, 1.00]), and more weakly with low oxygen saturations reflecting a larger right-to-left shunt (adjusted OR 0.96 [0.92, 1.01]). For the same pulmonary arteriovenous malformations, the stroke risk would approximately double with serum iron 6 µmol/L compared to mid-normal range (7–27 µmol/L). Platelet studies confirmed overlooked data that iron deficiency is associated with exuberant platelet aggregation to serotonin (5HT), correcting following iron treatment. By MANOVA, adjusting for participant and 5HT, iron or ferritin explained 14% of the variance in log-transformed aggregation-rate (p = 0.039/p = 0.021). Significance These data suggest that patients with compromised pulmonary capillary filtration due to pulmonary arteriovenous malformations are at increased risk of ischaemic stroke if they are iron deficient, and that mechanisms are likely to include enhanced aggregation of circulating platelets.
Collapse
|