1
|
Benavides-Nieto M, Adam F, Martin E, Boussard C, Lagresle-Peyrou C, Callebaut I, Kauskot A, Repérant C, Feng M, Bordet JC, Castelle M, Morelle G, Brouzes C, Zarhrate M, Panikulam P, Lambert N, Picard C, Bodet D, Rouger-Gaudichon J, Revy P, de Villartay JP, Moshous D. Somatic RAP1B gain-of-function variant underlies isolated thrombocytopenia and immunodeficiency. J Clin Invest 2024; 134:e169994. [PMID: 39225097 PMCID: PMC11364392 DOI: 10.1172/jci169994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/10/2024] [Indexed: 09/04/2024] Open
Abstract
The ubiquitously expressed small GTPase Ras-related protein 1B (RAP1B) acts as a molecular switch that regulates cell signaling, cytoskeletal remodeling, and cell trafficking and activates integrins in platelets and lymphocytes. The residue G12 in the P-loop is required for the RAP1B-GTPase conformational switch. Heterozygous germline RAP1B variants have been described in patients with syndromic thrombocytopenia. However, the causality and pathophysiological impact remained unexplored. We report a boy with neonatal thrombocytopenia, combined immunodeficiency, neutropenia, and monocytopenia caused by a heterozygous de novo single nucleotide substitution, c.35G>A (p.G12E) in RAP1B. We demonstrate that G12E and the previously described G12V and G60R were gain-of-function variants that increased RAP1B activation, talin recruitment, and integrin activation, thereby modifying late responses such as platelet activation, T cell proliferation, and migration. We show that in our patient, G12E was a somatic variant whose allele frequency decreased over time in the peripheral immune compartment, but remained stable in bone marrow cells, suggesting a differential effect in distinct cell populations. Allogeneic hematopoietic stem cell transplantation fully restored the patient's hemato-immunological phenotype. Our findings define monoallelic RAP1B gain-of-function variants as a cause for constitutive immunodeficiency and thrombocytopenia. The phenotypic spectrum ranged from isolated hematological manifestations in our patient with somatic mosaicism to complex syndromic features in patients with reported germline RAP1B variants.
Collapse
Affiliation(s)
- Marta Benavides-Nieto
- Université Paris Cité, Paris, France
- Imagine Institute, Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue Contre le Cancer, Ligue 2023, INSERM UMR 1163, Paris, France
- General Pediatrics–Infectious Diseases and Internal Medicine, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris (AP-HP) Nord, Paris, France
| | - Frédéric Adam
- INSERM UMR S 1176, Laboratory for Hemostasis, Inflammation and Thrombosis (HITh), Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Emmanuel Martin
- Laboratory Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Charlotte Boussard
- Université Paris Cité, Paris, France
- Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades University Hospital, AP-HP, Paris, France
- Laboratory Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Chantal Lagresle-Peyrou
- Biotherapy Clinical Investigation Center, AP-HP, Paris, France
- Laboratory Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France
| | - Isabelle Callebaut
- Sorbonne University, Muséum National d’Histoire Naturelle, UMR CNRS 7590, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, Paris, France
| | - Alexandre Kauskot
- INSERM UMR S 1176, Laboratory for Hemostasis, Inflammation and Thrombosis (HITh), Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Christelle Repérant
- INSERM UMR S 1176, Laboratory for Hemostasis, Inflammation and Thrombosis (HITh), Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Miao Feng
- INSERM UMR S 1176, Laboratory for Hemostasis, Inflammation and Thrombosis (HITh), Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Jean-Claude Bordet
- Laboratoire d’Hémostase, Centre de Biologie Est, Hospices Civils de Lyon, Bron, France
| | - Martin Castelle
- Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades University Hospital, AP-HP, Paris, France
| | - Guillaume Morelle
- Université Paris Cité, Paris, France
- Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades University Hospital, AP-HP, Paris, France
| | - Chantal Brouzes
- Laboratory of Onco-Hematology, Necker-Enfants Malades University Hospital, AP-HP, Paris, France, and INSERM U1151, Institut Necker-Enfants Malades, Paris, France
| | - Mohammed Zarhrate
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 and INSERM US24/CNRS UAR3633, Paris Descartes Sorbonne Paris Cité University, Paris, France
| | - Patricia Panikulam
- Université Paris Cité, Paris, France
- Laboratory “Molecular basis of altered immune homeostasis,” INSERM UMR 1163, Imagine Institute, Paris, France
| | - Nathalie Lambert
- Study Center for Primary Immunodeficiencies, Necker-Enfants Malades University Hospital, AP-HP, Paris, France
| | - Capucine Picard
- Université Paris Cité, Paris, France
- Laboratory Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Imagine Institute, Paris, France
- Study Center for Primary Immunodeficiencies, Necker-Enfants Malades University Hospital, AP-HP, Paris, France
- Centre de Référence des Déficits Immunitaires Héréditaires (CEREDIH), Necker-Enfants Malades University Hospital, AP-HP, Paris, France
| | - Damien Bodet
- CHU de Caen Normandie, Onco-Immunohématologie Pédiatrique, Caen, France
| | | | - Patrick Revy
- Université Paris Cité, Paris, France
- Imagine Institute, Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue Contre le Cancer, Ligue 2023, INSERM UMR 1163, Paris, France
| | - Jean-Pierre de Villartay
- Université Paris Cité, Paris, France
- Imagine Institute, Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue Contre le Cancer, Ligue 2023, INSERM UMR 1163, Paris, France
| | - Despina Moshous
- Université Paris Cité, Paris, France
- Imagine Institute, Laboratory of Genome Dynamics in the Immune System, Equipe Labellisée Ligue Contre le Cancer, Ligue 2023, INSERM UMR 1163, Paris, France
- Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades University Hospital, AP-HP, Paris, France
- Centre de Référence des Déficits Immunitaires Héréditaires (CEREDIH), Necker-Enfants Malades University Hospital, AP-HP, Paris, France
| |
Collapse
|
2
|
Paul S, Mukherjee T, Das K. Coagulation Protease-Driven Cancer Immune Evasion: Potential Targets for Cancer Immunotherapy. Cancers (Basel) 2024; 16:1568. [PMID: 38672649 PMCID: PMC11048528 DOI: 10.3390/cancers16081568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Blood coagulation and cancer are intrinsically connected, hypercoagulation-associated thrombotic complications are commonly observed in certain types of cancer, often leading to decreased survival in cancer patients. Apart from the common role in coagulation, coagulation proteases often trigger intracellular signaling in various cancers via the activation of a G protein-coupled receptor superfamily protease: protease-activated receptors (PARs). Although the role of PARs is well-established in the development and progression of certain types of cancer, their impact on cancer immune response is only just emerging. The present review highlights how coagulation protease-driven PAR signaling plays a key role in modulating innate and adaptive immune responses. This is followed by a detailed discussion on the contribution of coagulation protease-induced signaling in cancer immune evasion, thereby supporting the growth and development of certain tumors. A special section of the review demonstrates the role of coagulation proteases, thrombin, factor VIIa, and factor Xa in cancer immune evasion. Targeting coagulation protease-induced signaling might be a potential therapeutic strategy to boost the immune surveillance mechanism of a host fighting against cancer, thereby augmenting the clinical consequences of targeted immunotherapeutic regimens.
Collapse
Affiliation(s)
- Subhojit Paul
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, West Bengal, India;
| | - Tanmoy Mukherjee
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA;
| | - Kaushik Das
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani 741251, West Bengal, India
| |
Collapse
|
3
|
Kauskot A, Mallebranche C, Bruneel A, Fenaille F, Solarz J, Viellard T, Feng M, Repérant C, Bordet JC, Cholet S, Denis CV, McCluskey G, Latour S, Martin E, Pellier I, Lasne D, Borgel D, Kracker S, Ziegler A, Tuffigo M, Fournier B, Miot C, Adam F. MAGT1 deficiency in XMEN disease is associated with severe platelet dysfunction and impaired platelet glycoprotein N-glycosylation. J Thromb Haemost 2023; 21:3268-3278. [PMID: 37207862 DOI: 10.1016/j.jtha.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/24/2023] [Accepted: 05/07/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND X-linked immunodeficiency with magnesium defect, Epstein-Barr virus infection, and neoplasia (XMEN) disease is a primary immunodeficiency due to loss-of-function mutations in the gene encoding for magnesium transporter 1 (MAGT1). Furthermore, as MAGT1 is involved in the N-glycosylation process, XMEN disease is classified as a congenital disorder of glycosylation. Although XMEN-associated immunodeficiency is well described, the mechanisms underlying platelet dysfunction and those responsible for life-threatening bleeding events have never been investigated. OBJECTIVES To assess platelet functions in patients with XMEN disease. METHODS Two unrelated young boys, including one before and after hematopoietic stem cell transplantation, were investigated for their platelet functions, glycoprotein expression, and serum and platelet-derived N-glycans. RESULTS Platelet analysis highlighted abnormal elongated cells and unusual barbell-shaped proplatelets. Platelet aggregation, integrin αIIbβ3 activation, calcium mobilization, and protein kinase C activity were impaired between both patients. Strikingly, platelet responses to protease-activated receptor 1 activating peptide were absent at both low and high concentrations. These defects were also associated with decreased molecular weights of glycoprotein Ibα, glycoprotein VI, and integrin αIIb due to partial impairment of N-glycosylation. All these defects were corrected after hematopoietic stem cell transplantation. CONCLUSION Our results highlight prominent platelet dysfunction related to MAGT1 deficiency and defective N-glycosylation in several platelet proteins that could explain the hemorrhages reported in patients with XMEN disease.
Collapse
Affiliation(s)
- Alexandre Kauskot
- INSERM U1176, Hemostasis, Inflammation & Thrombosis (HITh), Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Coralie Mallebranche
- Université d'Angers, Université de Nantes, Inserm, CNRS, CRCI2NA, SFR ICAT, Angers, France; CHU Angers, Pediatric immuno-hemato-oncology Unit, Angers, France
| | - Arnaud Bruneel
- AP-HP, Biochimie Métabolique et Cellulaire, Hôpital Bichat-Claude Bernard, Paris, France; Université Paris-Saclay, INSERM UMR1193, Mécanismes cellulaires et moléculaires de l'adaptation au stress et cancérogenèse, Châtenay-Malabry, France
| | - François Fenaille
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, MetaboHUB, Gif sur Yvette, France
| | - Jean Solarz
- INSERM U1176, Hemostasis, Inflammation & Thrombosis (HITh), Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Toscane Viellard
- INSERM U1176, Hemostasis, Inflammation & Thrombosis (HITh), Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Miao Feng
- INSERM U1176, Hemostasis, Inflammation & Thrombosis (HITh), Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Christelle Repérant
- INSERM U1176, Hemostasis, Inflammation & Thrombosis (HITh), Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Jean-Claude Bordet
- Laboratoire d'Hémostase, Centre de Biologie Est, Hospices Civils de Lyon, Bron, France
| | - Sophie Cholet
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, MetaboHUB, Gif sur Yvette, France
| | - Cécile V Denis
- INSERM U1176, Hemostasis, Inflammation & Thrombosis (HITh), Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Geneviève McCluskey
- INSERM U1176, Hemostasis, Inflammation & Thrombosis (HITh), Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Sylvain Latour
- INSERM UMR 1163, Laboratory of Lymphocyte Activation and Susceptibility to EBV, Imagine Institute, Université Paris Cité, Paris, France
| | - Emmanuel Martin
- INSERM UMR 1163, Laboratory of Lymphocyte Activation and Susceptibility to EBV, Imagine Institute, Université Paris Cité, Paris, France
| | - Isabelle Pellier
- Université d'Angers, Université de Nantes, Inserm, CNRS, CRCI2NA, SFR ICAT, Angers, France; CHU Angers, Pediatric immuno-hemato-oncology Unit, Angers, France
| | - Dominique Lasne
- INSERM U1176, Hemostasis, Inflammation & Thrombosis (HITh), Université Paris-Saclay, Le Kremlin-Bicêtre, France; AP-HP, Laboratoire d'Hématologie, Hôpital Necker-Enfants Malades, Paris, France
| | - Delphine Borgel
- INSERM U1176, Hemostasis, Inflammation & Thrombosis (HITh), Université Paris-Saclay, Le Kremlin-Bicêtre, France; AP-HP, Laboratoire d'Hématologie, Hôpital Necker-Enfants Malades, Paris, France
| | - Sven Kracker
- INSERM UMR1163, Université Paris Cité, Laboratory of Human Lymphohematopoiesis, Imagine Institute, Paris, France
| | | | - Marie Tuffigo
- CHU Angers, Laboratory of Hematology, Angers, France
| | - Benjamin Fournier
- INSERM UMR 1163, Laboratory of Lymphocyte Activation and Susceptibility to EBV, Imagine Institute, Université Paris Cité, Paris, France; AP-HP, Hôpital Necker-Enfants Malades Assistance Publique-Hôpitaux de Paris, Pediatric Hematology-Immunology-Rheumatology Unit, Paris, France
| | - Charline Miot
- Université d'Angers, Université de Nantes, Inserm, CNRS, CRCI2NA, SFR ICAT, Angers, France; CHU Angers, Pediatric immuno-hemato-oncology Unit, Angers, France; CHU Angers, Laboratory of Immunology and Allergology, Angers, France
| | - Frédéric Adam
- INSERM U1176, Hemostasis, Inflammation & Thrombosis (HITh), Université Paris-Saclay, Le Kremlin-Bicêtre, France.
| |
Collapse
|
4
|
Oshinowo O, Azer SS, Lin J, Lam WA. Why platelet mechanotransduction matters for hemostasis and thrombosis. J Thromb Haemost 2023; 21:2339-2353. [PMID: 37331517 PMCID: PMC10529432 DOI: 10.1016/j.jtha.2023.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 06/20/2023]
Abstract
Mechanotransduction is the ability of cells to "feel" or sense their mechanical microenvironment and integrate and convert these physical stimuli into adaptive biochemical cellular responses. This phenomenon is vital for the physiology of numerous nucleated cell types to affect their various cellular processes. As the main drivers of hemostasis and clot retraction, platelets also possess this ability to sense the dynamic mechanical microenvironments of circulation and convert those signals into biological responses integral to clot formation. Like other cell types, platelets leverage their "hands" or receptors/integrins to mechanotransduce important signals in responding to vascular injury to achieve hemostasis. The clinical relevance of cellular mechanics and mechanotransduction is imperative as pathologic alterations or aberrant mechanotransduction in platelets has been shown to lead to bleeding and thrombosis. As such, the aim of this review is to provide an overview of the most recent research related to platelet mechanotransduction, from platelet generation to platelet activation, within the hemodynamic environment and clot contraction at the site of vascular injury, thereby covering the entire "life cycle" of platelets. Additionally, we describe the key mechanoreceptors in platelets and discuss the new biophysical techniques that have enabled the field to understand how platelets sense and respond to their mechanical microenvironment via those receptors. Finally, the clinical significance and importance of continued exploration of platelet mechanotransduction have been discussed as the key to better understanding of both thrombotic and bleeding disorders lies in a more complete mechanistic understanding of platelet function by way of mechanotransduction.
Collapse
Affiliation(s)
- Oluwamayokun Oshinowo
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA; The Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, Georgia, USA; Children's Healthcare of Atlanta Inc, Aflac Cancer and Blood Disorders Center, Atlanta, Georgia, USA
| | - Sally S Azer
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA; The Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, Georgia, USA; Children's Healthcare of Atlanta Inc, Aflac Cancer and Blood Disorders Center, Atlanta, Georgia, USA
| | - Jessica Lin
- The Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Wilbur A Lam
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA; The Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, Georgia, USA; Children's Healthcare of Atlanta Inc, Aflac Cancer and Blood Disorders Center, Atlanta, Georgia, USA.
| |
Collapse
|
5
|
Knauss EA, Nieman MT. Glycoprotein V exerts local control of fibrin. NATURE CARDIOVASCULAR RESEARCH 2023; 2:335-336. [PMID: 39195957 DOI: 10.1038/s44161-023-00261-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Elizabeth A Knauss
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Marvin T Nieman
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
6
|
Lin YT, Li Y, Hsu HC, Tsai JY, Lee JH, Tai CJ, Wu MJ, Wu CC. Discovery of 7, 4'-dimethoxy-3-hydroxyflavone as a protease-activated receptor 4 antagonist with antithrombotic activity and less bleeding tendency in mice. Biochem Pharmacol 2022; 202:115152. [PMID: 35752281 DOI: 10.1016/j.bcp.2022.115152] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/31/2022] [Accepted: 06/17/2022] [Indexed: 11/29/2022]
Abstract
There is growing evidence of the importance of protease-activated receptor 4 (PAR4), one of thrombin receptors, as a therapeutic target in thrombotic cardiovascular diseases. In the present study, we utilized ligand-based virtual screening, bioassay, and structure-activity relationship study to discover PAR4 antagonists with new chemical scaffolds from natural origin, and examined their application as antiplatelet agents. By using these approaches, we have identified a flavonoid, 7, 4'-dimethoxy-3-hydroxyflavone, that exhibits anti-PAR4 activity. 7, 4'-Dimethoxy-3-hydroxyflavone inhibited PAR4-mediated human platelet aggregation, GPIIb/IIIa activation, and P-selectin secretion. Also, it inhibited PAR4 downstream signaling pathways, including Ca2+/protein kinase C, Akt, and MAP kinases ERK and p38, in human platelets, and suppressed PAR4-mediated β-arrestin recruitment in CHO-K1 cells exogenously expressed human PAR4. In a microfluidic system, 7, 4'-dimethoxy-3-hydroxyflavone reduced thrombus formation on collagen-coated chambers at an arterial shear rate in recalcified whole blood. Furthermore, mice treated with 7, 4'-dimethoxy-3-hydroxyflavone were significantly protected from FeCl3-induced carotid arterial occlusions, without significantly affecting tail bleeding time. In conclusion, 7, 4'-dimethoxy-3-hydroxyflavone represents a new class of nature-based PAR4 antagonist, it shows effective in vivo antithrombotic properties with less bleeding tendency, and could be a potential candidate for developing new antiplatelet agents.
Collapse
Affiliation(s)
- Ying-Ting Lin
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yu Li
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Hui-Ching Hsu
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ju-Ying Tsai
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Jia-Hau Lee
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chi-Jung Tai
- Department of Family Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ming-Jung Wu
- Department of Chemistry, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Chin-Chung Wu
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
7
|
Gnanenthiran SR, Pennings GJ, Reddel CJ, Campbell H, Kockx M, Hamilton JR, Chen V, Kritharides L. Identification of a Distinct Platelet Phenotype in the Elderly: ADP Hypersensitivity Coexists With Platelet PAR (Protease-Activated Receptor)-1 and PAR-4-Mediated Thrombin Resistance. Arterioscler Thromb Vasc Biol 2022; 42:960-972. [PMID: 35708029 DOI: 10.1161/atvbaha.120.316772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Thrombin (via PAR [protease-activated receptor]-1 and PAR-4) and ADP (via P2Y12 receptors) are potent endogenous platelet activators implicated in the development of cardiovascular disease. We aimed to assess whether platelet pathways alter with aging. METHODS We characterized platelet activity in community-dwelling volunteers (n=174) in the following age groups: (1) 20 to 30 (young); (2) 40 to 55 (middle-aged); (3) ≥70 years (elderly). Platelet activity was assessed by aggregometry; flow cytometry (surface markers [P-selectin: alpha granule release, CD63: dense granule release, PAC-1 (measure of conformationally active GPIIb/IIIa at the fibrinogen binding site): GPIIb/IIIa conformational activation] measured under basal conditions and after agonist stimulation [ADP, thrombin, PAR-1 agonist or PAR-4 agonist]); receptor cleavage and quantification; fluorometry; calcium flux; ELISA. RESULTS The elderly had higher basal platelet activation than the young, evidenced by increased expression of P-selectin, CD63, and PAC-1, which correlated with increasing inflammation (IL [interleukin]-1β/IL-6). The elderly demonstrated higher P2Y12 receptor density, with greater ADP-induced platelet aggregation (P<0.05). However, elderly subjects were resistant to thrombin, achieving less activation in response to thrombin (higher EC50) and to selective stimulation of both PAR-1 and PAR-4, with higher basal PAR-1/PAR-4 cleavage and less inducible PAR-1/PAR-4 cleavage (all P<0.05). Thrombin resistance was attributable to a combination of reduced thrombin orienting receptor GPIbα, reduced secondary ADP contribution to thrombin-mediated activation, and blunted calcium flux. D-Dimer, a marker of in situ thrombin generation, correlated with platelet activation in the circulation, ex vivo thrombin resistance, and circulating inflammatory mediators (TNF [tumor necrosis factor]-α/IL-6). CONCLUSIONS Aging is associated with a distinctive platelet phenotype of increased basal activation, ADP hyperreactivity, and thrombin resistance. In situ thrombin generation associated with systemic inflammation may be novel target to prevent cardiovascular disease in the elderly.
Collapse
Affiliation(s)
- Sonali R Gnanenthiran
- Cardiology Department, Concord Repatriation General Hospital, NSW, Australia (S.R.G., M.K., L.K.).,ANZAC Research Institute, Concord Repatriation General Hospital, and University of Sydney, Sydney, NSW, Australia (S.R.G., G.J.P., C.J.R., H.C., M.K., V.C., L.K.)
| | - Gabrielle J Pennings
- ANZAC Research Institute, Concord Repatriation General Hospital, and University of Sydney, Sydney, NSW, Australia (S.R.G., G.J.P., C.J.R., H.C., M.K., V.C., L.K.)
| | - Caroline J Reddel
- ANZAC Research Institute, Concord Repatriation General Hospital, and University of Sydney, Sydney, NSW, Australia (S.R.G., G.J.P., C.J.R., H.C., M.K., V.C., L.K.)
| | - Heather Campbell
- ANZAC Research Institute, Concord Repatriation General Hospital, and University of Sydney, Sydney, NSW, Australia (S.R.G., G.J.P., C.J.R., H.C., M.K., V.C., L.K.)
| | - Maaike Kockx
- Cardiology Department, Concord Repatriation General Hospital, NSW, Australia (S.R.G., M.K., L.K.).,ANZAC Research Institute, Concord Repatriation General Hospital, and University of Sydney, Sydney, NSW, Australia (S.R.G., G.J.P., C.J.R., H.C., M.K., V.C., L.K.)
| | - Justin R Hamilton
- Australian Centre of Blood Diseases, Monash University, Victoria, Australia (J.R.H.)
| | - Vivien Chen
- ANZAC Research Institute, Concord Repatriation General Hospital, and University of Sydney, Sydney, NSW, Australia (S.R.G., G.J.P., C.J.R., H.C., M.K., V.C., L.K.).,Haematology Department, Concord Repatriation General Hospital, NSW, Australia (V.C.)
| | - Leonard Kritharides
- Cardiology Department, Concord Repatriation General Hospital, NSW, Australia (S.R.G., M.K., L.K.).,ANZAC Research Institute, Concord Repatriation General Hospital, and University of Sydney, Sydney, NSW, Australia (S.R.G., G.J.P., C.J.R., H.C., M.K., V.C., L.K.)
| |
Collapse
|
8
|
Abstract
During sepsis, an initial prothrombotic shift takes place, in which coagulatory acute-phase proteins are increased, while anticoagulatory factors and platelet count decrease. Further on, the fibrinolytic system becomes impaired, which contributes to disease severity. At a later stage in sepsis, coagulation factors may become depleted, and sepsis patients may shift into a hypo-coagulable state with an increased bleeding risk. During the pro-coagulatory shift, critically ill patients have an increased thrombosis risk that ranges from developing micro-thromboses that impair organ function to life-threatening thromboembolic events. Here, thrombin plays a key role in coagulation as well as in inflammation. For thromboprophylaxis, low molecular weight heparins (LMWH) and unfractionated heparins (UFHs) are recommended. Nevertheless, there are conditions such as heparin resistance or heparin-induced thrombocytopenia (HIT), wherein heparin becomes ineffective or even puts the patient at an increased prothrombotic risk. In these cases, argatroban, a direct thrombin inhibitor (DTI), might be a potential alternative anticoagulatory strategy. Yet, caution is advised with regard to dosing of argatroban especially in sepsis. Therefore, the starting dose of argatroban is recommended to be low and should be titrated to the targeted anticoagulation level and be closely monitored in the further course of treatment. The authors of this review recommend using DTIs such as argatroban as an alternative anticoagulant in critically ill patients suffering from sepsis or COVID-19 with suspected or confirmed HIT, HIT-like conditions, impaired fibrinolysis, in patients on extracorporeal circuits and patients with heparin resistance, when closely monitored.
Collapse
|
9
|
Bendas G, Schlesinger M. The GPIb-IX complex on platelets: insight into its novel physiological functions affecting immune surveillance, hepatic thrombopoietin generation, platelet clearance and its relevance for cancer development and metastasis. Exp Hematol Oncol 2022; 11:19. [PMID: 35366951 PMCID: PMC8976409 DOI: 10.1186/s40164-022-00273-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/19/2022] [Indexed: 12/13/2022] Open
Abstract
The glycoprotein (GP) Ib-IX complex is a platelet receptor that mediates the initial interaction with subendothelial von Willebrand factor (VWF) causing platelet arrest at sites of vascular injury even under conditions of high shear. GPIb-IX dysfunction or deficiency is the reason for the rare but severe Bernard-Soulier syndrome (BSS), a congenital bleeding disorder. Although knowledge on GPIb-IX structure, its basic functions, ligands, and intracellular signaling cascades have been well established, several advances in GPIb-IX biology have been made in the recent years. Thus, two mechanosensitive domains and a trigger sequence in GPIb were characterized and its role as a thrombin receptor was deciphered. Furthermore, it became clear that GPIb-IX is involved in the regulation of platelet production, clearance and thrombopoietin secretion. GPIb is deemed to contribute to liver cancer development and metastasis. This review recapitulates these novel findings highlighting GPIb-IX in its multiple functions as a key for immune regulation, host defense, and liver cancer development.
Collapse
Affiliation(s)
- Gerd Bendas
- Department of Pharmacy, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Martin Schlesinger
- Department of Pharmacy, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121, Bonn, Germany. .,Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany.
| |
Collapse
|
10
|
Long-term safety and efficacy of lentiviral hematopoietic stem/progenitor cell gene therapy for Wiskott-Aldrich syndrome. Nat Med 2022; 28:71-80. [PMID: 35075289 PMCID: PMC8799465 DOI: 10.1038/s41591-021-01641-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 11/24/2021] [Indexed: 12/14/2022]
Abstract
Patients with Wiskott–Aldrich syndrome (WAS) lacking a human leukocyte antigen-matched donor may benefit from gene therapy through the provision of gene-corrected, autologous hematopoietic stem/progenitor cells. Here, we present comprehensive, long-term follow-up results (median follow-up, 7.6 years) (phase I/II trial no. NCT02333760) for eight patients with WAS having undergone phase I/II lentiviral vector-based gene therapy trials (nos. NCT01347346 and NCT01347242), with a focus on thrombocytopenia and autoimmunity. Primary outcomes of the long-term study were to establish clinical and biological safety, efficacy and tolerability by evaluating the incidence and type of serious adverse events and clinical status and biological parameters including lentiviral genomic integration sites in different cell subpopulations from 3 years to 15 years after gene therapy. Secondary outcomes included monitoring the need for additional treatment and T cell repertoire diversity. An interim analysis shows that the study meets the primary outcome criteria tested given that the gene-corrected cells engrafted stably, and no serious treatment-associated adverse events occurred. Overall, severe infections and eczema resolved. Autoimmune disorders and bleeding episodes were significantly less frequent, despite only partial correction of the platelet compartment. The results suggest that lentiviral gene therapy provides sustained clinical benefits for patients with WAS. Long-term monitoring of patients with Wiskott–Aldrich syndrome following lentiviral gene therapy shows a safe profile and a reduction in the frequency of autoimmune manifestations and bleeding events, despite incomplete platelet reconstitution.
Collapse
|
11
|
Razanakolona M, Adam F, Bianchini E, Saller F, Carvalho AD, Diehl JL, Denis CV, Meziani F, Borgel D, Helms J, Vasse M. Anti-inflammatory Activity of the Protein Z-Dependent Protease Inhibitor. TH OPEN 2021; 5:e220-e229. [PMID: 34189397 PMCID: PMC8233056 DOI: 10.1055/s-0041-1730037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 04/08/2021] [Indexed: 12/19/2022] Open
Abstract
The protein Z (PZ)-dependent plasma protease inhibitor (ZPI) is a glycoprotein that inhibits factor XIa and, in the presence of PZ, FXa. Recently, ZPI has been shown to be an acute-phase protein (APP). As usually APPs downregulate the harmful effects of inflammation, we tested whether ZPI could modulate the increase of cytokines observed in inflammatory states. We observed that recombinant human ZPI (rhZPI) significantly decreases the levels of interleukin (IL)-1, IL-6, and tumor necrosis factor- α (TNF-α) induced by lipopolysaccharide (LPS) in a whole blood model. This inhibitory effect was unaffected by the presence of PZ or heparin. A ZPI mutant within the reactive loop center ZPI (Y387A), lacking anticoagulant activity, still had an anti-inflammatory activity. Surprisingly, rhZPI did not inhibit the synthesis of IL-6 or TNF-α when purified monocytes were stimulated by LPS, whereas the inhibitory effect was evidenced when lymphocytes were added to monocytes. The requirement of lymphocytes could be due to the synthesis of CCL5 (RANTES), a chemokine mainly produced by activated lymphocytes which is induced by rhZPI, and which can reduce the production of proinflammatory cytokines in whole blood. Lastly, we observed that the intraperitoneal injection of rhZPI significantly decreased LPS-induced IL-6 and TNF-α production in mouse plasma.
Collapse
Affiliation(s)
- Mahita Razanakolona
- HITh, INSERM, UMR_S1176, Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France
| | - Frédéric Adam
- HITh, INSERM, UMR_S1176, Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France
| | - Elsa Bianchini
- HITh, INSERM, UMR_S1176, Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France
| | - François Saller
- HITh, INSERM, UMR_S1176, Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France
| | - Allan de Carvalho
- HITh, INSERM, UMR_S1176, Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France
| | - Jean-Luc Diehl
- Département de réanimation médicale, Hôpital Européen Georges Pompidou, Paris, France
| | - Cécile V Denis
- HITh, INSERM, UMR_S1176, Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France
| | - Ferhat Meziani
- Faculté de Médecine, Service de Médecine Intensive-Réanimation, Université de Strasbourg (UNISTRA), Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, Strasbourg, France.,INSERM (French National Institute of Health and Medical Research), Regenerative Nanomedicine (RNM), FMTS, Strasbourg, France
| | - Delphine Borgel
- HITh, INSERM, UMR_S1176, Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France.,APHP, Laboratoire d'Hématologie, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Julie Helms
- Faculté de Médecine, Service de Médecine Intensive-Réanimation, Université de Strasbourg (UNISTRA), Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, Strasbourg, France.,ImmunoRhumatologie Moléculaire, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Marc Vasse
- HITh, INSERM, UMR_S1176, Université Paris-Saclay, Le Kremlin-Bicêtre cedex, France.,Service de Biologie Clinique, Hôpital Foch, Suresnes, France
| |
Collapse
|
12
|
Zwifelhofer NMJ, Bercovitz RS, Cole R, Yan K, Simpson PM, Moroi A, Newman PJ, Niebler RA, Scott JP, Stuth EAD, Woods RK, Benson DW, Newman DK. Platelet Function Changes during Neonatal Cardiopulmonary Bypass Surgery: Mechanistic Basis and Lack of Correlation with Excessive Bleeding. Thromb Haemost 2019; 120:94-106. [PMID: 31752040 DOI: 10.1055/s-0039-1700517] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Thrombocytopenia and platelet dysfunction induced by extracorporeal blood circulation are thought to contribute to postsurgical bleeding complications in neonates undergoing cardiac surgery with cardiopulmonary bypass (CPB). In this study, we examined how changes in platelet function relate to changes in platelet count and to excessive bleeding in neonatal CPB surgery. Platelet counts and platelet P-selectin exposure in response to agonist stimulation were measured at four times before, during, and after CPB surgery in neonates with normal versus excessive levels of postsurgical bleeding. Relative to baseline, platelet counts were reduced in patients while on CPB, as was platelet activation by the thromboxane A2 analog U46619, thrombin receptor activating peptide (TRAP), and collagen-related peptide (CRP). Platelet activation by adenosine diphosphate (ADP) was instead reduced after platelet transfusion. We provide evidence that thrombocytopenia is a likely contributor to CPB-associated defects in platelet responsiveness to U46619 and TRAP, CPB-induced collagen receptor downregulation likely contributes to defective platelet responsiveness to CRP, and platelet transfusion may contribute to defective platelet responses to ADP. Platelet transfusion restored to baseline levels platelet counts and responsiveness to all agonists except ADP but did not prevent excessive bleeding in all patients. We conclude that platelet count and function defects are characteristic of neonatal CPB surgery and that platelet transfusion corrects these defects. However, since CPB-associated coagulopathy is multifactorial, platelet transfusion alone is insufficient to treat bleeding events in all patients. Therefore, platelet transfusion must be combined with treatment of other factors that contribute to the coagulopathy to prevent excessive bleeding.
Collapse
Affiliation(s)
| | - Rachel S Bercovitz
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Regina Cole
- Herma Heart Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin, United States.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Ke Yan
- Herma Heart Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin, United States.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Pippa M Simpson
- Herma Heart Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin, United States.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Alyssa Moroi
- Versiti-Blood Research Institute, Milwaukee, Wisconsin, United States
| | - Peter J Newman
- Versiti-Blood Research Institute, Milwaukee, Wisconsin, United States.,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Robert A Niebler
- Herma Heart Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin, United States.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - John P Scott
- Herma Heart Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin, United States.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States.,Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Eckehard A D Stuth
- Herma Heart Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin, United States.,Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Ronald K Woods
- Herma Heart Institute, Children's Hospital of Wisconsin, Milwaukee, Wisconsin, United States.,Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - D Woodrow Benson
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Debra K Newman
- Versiti-Blood Research Institute, Milwaukee, Wisconsin, United States.,Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
13
|
Stoupa A, Adam F, Kariyawasam D, Strassel C, Gawade S, Szinnai G, Kauskot A, Lasne D, Janke C, Natarajan K, Schmitt A, Bole-Feysot C, Nitschke P, Léger J, Jabot-Hanin F, Tores F, Michel A, Munnich A, Besmond C, Scharfmann R, Lanza F, Borgel D, Polak M, Carré A. TUBB1 mutations cause thyroid dysgenesis associated with abnormal platelet physiology. EMBO Mol Med 2019; 10:emmm.201809569. [PMID: 30446499 PMCID: PMC6284387 DOI: 10.15252/emmm.201809569] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The genetic causes of congenital hypothyroidism due to thyroid dysgenesis (TD) remain largely unknown. We identified three novel TUBB1 gene mutations that co‐segregated with TD in three distinct families leading to 1.1% of TUBB1 mutations in TD study cohort. TUBB1 (Tubulin, Beta 1 Class VI) encodes for a member of the β‐tubulin protein family. TUBB1 gene is expressed in the developing and adult thyroid in humans and mice. All three TUBB1 mutations lead to non‐functional α/β‐tubulin dimers that cannot be incorporated into microtubules. In mice, Tubb1 knock‐out disrupted microtubule integrity by preventing β1‐tubulin incorporation and impaired thyroid migration and thyroid hormone secretion. In addition, TUBB1 mutations caused the formation of macroplatelets and hyperaggregation of human platelets after stimulation by low doses of agonists. Our data highlight unexpected roles for β1‐tubulin in thyroid development and in platelet physiology. Finally, these findings expand the spectrum of the rare paediatric diseases related to mutations in tubulin‐coding genes and provide new insights into the genetic background and mechanisms involved in congenital hypothyroidism and thyroid dysgenesis.
Collapse
Affiliation(s)
- Athanasia Stoupa
- INSERM U1016, Faculté de Médecine, Cochin Institute, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,IMAGINE Institute Affiliate, Paris, France.,RARE Disorder Center: Centre des Maladies Endocriniennes Rares de la Croissance et du Développement, Paris, France.,Pediatric Endocrinology, Gynecology and Diabetology Unit, Hôpital Universitaire Necker-Enfants Malades, AP-HP, Paris, France
| | - Frédéric Adam
- INSERM UMR_S1176, Paris-Sud University, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Dulanjalee Kariyawasam
- RARE Disorder Center: Centre des Maladies Endocriniennes Rares de la Croissance et du Développement, Paris, France.,Pediatric Endocrinology, Gynecology and Diabetology Unit, Hôpital Universitaire Necker-Enfants Malades, AP-HP, Paris, France
| | - Catherine Strassel
- INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Université de Strasbourg, Strasbourg, France
| | - Sanjay Gawade
- Department of Biomedicine, Pediatric Immunology, University of Basel, Basel, Switzerland
| | - Gabor Szinnai
- Department of Biomedicine, Pediatric Immunology, University of Basel, Basel, Switzerland.,Pediatric Endocrinology, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Alexandre Kauskot
- INSERM UMR_S1176, Paris-Sud University, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Dominique Lasne
- INSERM UMR_S1176, Paris-Sud University, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Necker Children's Hospital, Biological Hematology Service, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Carsten Janke
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France.,Institut Curie, CNRS UMR3348, Université Paris Sud, Université Paris-Saclay, Orsay, France
| | - Kathiresan Natarajan
- Institut Curie, CNRS UMR3348, PSL Research University, Orsay, France.,Institut Curie, CNRS UMR3348, Université Paris Sud, Université Paris-Saclay, Orsay, France
| | - Alain Schmitt
- INSERM U1016, Faculté de Médecine, Cochin Institute, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Christine Bole-Feysot
- Genomic Platform, INSERM UMR 1163, IMAGINE Institute, Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Patrick Nitschke
- Bioinformatics Platform, IMAGINE Institute, Paris Descartes University, Paris, France
| | - Juliane Léger
- RARE Disorder Center: Centre des Maladies Endocriniennes Rares de la Croissance et du Développement, Paris, France.,Pediatric Endocrinology Unit, Hôpital Universitaire Robert Debré, AP-HP, Paris, France.,Paris Diderot University, Sorbonne Paris Cité, Paris, France.,INSERM UMR 1141, DHU Protect, Paris, France
| | - Fabienne Jabot-Hanin
- Bioinformatics Platform, IMAGINE Institute, Paris Descartes University, Paris, France
| | - Frédéric Tores
- Bioinformatics Platform, IMAGINE Institute, Paris Descartes University, Paris, France
| | - Anita Michel
- INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Université de Strasbourg, Strasbourg, France
| | - Arnold Munnich
- INSERM U1163, IMAGINE Institute, Translational Genetics, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Department of Genetics, Hôpital Universitaire Necker-Enfants Malades, AP-HP, Paris, France
| | - Claude Besmond
- INSERM U1163, IMAGINE Institute, Translational Genetics, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Raphaël Scharfmann
- INSERM U1016, Faculté de Médecine, Cochin Institute, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - François Lanza
- INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Université de Strasbourg, Strasbourg, France
| | - Delphine Borgel
- INSERM UMR_S1176, Paris-Sud University, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Necker Children's Hospital, Biological Hematology Service, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Michel Polak
- INSERM U1016, Faculté de Médecine, Cochin Institute, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,IMAGINE Institute Affiliate, Paris, France.,RARE Disorder Center: Centre des Maladies Endocriniennes Rares de la Croissance et du Développement, Paris, France.,Pediatric Endocrinology, Gynecology and Diabetology Unit, Hôpital Universitaire Necker-Enfants Malades, AP-HP, Paris, France.,Fédération Parisienne pour le Dépistage et la Prévention des Handicaps de l'Enfant (FPDPHE), Paris, France
| | - Aurore Carré
- INSERM U1016, Faculté de Médecine, Cochin Institute, Université Paris Descartes, Sorbonne Paris Cité, Paris, France .,IMAGINE Institute Affiliate, Paris, France.,RARE Disorder Center: Centre des Maladies Endocriniennes Rares de la Croissance et du Développement, Paris, France
| |
Collapse
|
14
|
|
15
|
Adam F, Kauskot A, Kurowska M, Goudin N, Munoz I, Bordet JC, Huang JD, Bryckaert M, Fischer A, Borgel D, de Saint Basile G, Christophe OD, Ménasché G. Kinesin-1 Is a New Actor Involved in Platelet Secretion and Thrombus Stability. Arterioscler Thromb Vasc Biol 2018. [PMID: 29519941 DOI: 10.1161/atvbaha.117.310373] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Platelet secretion is crucial for many physiological platelet responses. Even though several regulators of the fusion machinery for secretory granule exocytosis have been identified in platelets, the underlying mechanisms are not yet fully characterized. APPROACH AND RESULTS By studying a mouse model (cKO [conditional knockout]Kif5b) lacking Kif5b (kinesin-1 heavy chain) in its megakaryocytes and platelets, we evidenced unstable hemostasis characterized by an increase of blood loss associated to a marked tendency to rebleed in a tail-clip assay and thrombus instability in an in vivo thrombosis model. This instability was confirmed in vitro in a whole-blood perfusion assay under blood flow conditions. Aggregations induced by thrombin and collagen were also impaired in cKOKif5b platelets. Furthermore, P-selectin exposure, PF4 (platelet factor 4) secretion, and ATP release after thrombin stimulation were impaired in cKOKif5b platelets, highlighting the role of kinesin-1 in α-granule and dense granule secretion. Importantly, exogenous ADP rescued normal thrombin induced-aggregation in cKOKif5b platelets, which indicates that impaired aggregation was because of defective release of ADP and dense granules. Last, we demonstrated that kinesin-1 interacts with the molecular machinery comprising the granule-associated Rab27 (Ras-related protein Rab-27) protein and the Slp4 (synaptotagmin-like protein 4/SYTL4) adaptor protein. CONCLUSIONS Our results indicate that a kinesin-1-dependent process plays a role for platelet function by acting into the mechanism underlying α-granule and dense granule secretion.
Collapse
Affiliation(s)
- Frédéric Adam
- From the INSERM, UMR_S 1176, Paris-Sud University, Université Paris-Saclay, Le Kremlin-Bicêtre, France (F.A., A.K., M.B., D.B., O.D.C.)
| | - Alexandre Kauskot
- From the INSERM, UMR_S 1176, Paris-Sud University, Université Paris-Saclay, Le Kremlin-Bicêtre, France (F.A., A.K., M.B., D.B., O.D.C.)
| | - Mathieu Kurowska
- INSERM, UMR_S 1163, Laboratory of Normal and Pathological Homeostasis of the Immune System, Paris, France (M.K., I.M., A.F., G.d.S.B., G.M.).,Imagine Institute (M.K., I.M., A.F., G.d.S.B., G.M.)
| | - Nicolas Goudin
- Cell Imaging Facility, Imagine Institute (N.G.), Paris Descartes University, Sorbonne Paris Cité, France
| | - Isabelle Munoz
- INSERM, UMR_S 1163, Laboratory of Normal and Pathological Homeostasis of the Immune System, Paris, France (M.K., I.M., A.F., G.d.S.B., G.M.).,Imagine Institute (M.K., I.M., A.F., G.d.S.B., G.M.)
| | - Jean-Claude Bordet
- Laboratoire d'Hémostase, Centre de Biologie Est, Hospices Civils de Lyon, Bron, France (J.-C.B.).,Laboratoire de Recherche sur l'Hémophilie, UCBL1, Lyon, France (J.-C.B.)
| | - Jian-Dong Huang
- School of Biomedical Sciences, The University of Hong Kong, China (J.-D.H.)
| | - Marijke Bryckaert
- From the INSERM, UMR_S 1176, Paris-Sud University, Université Paris-Saclay, Le Kremlin-Bicêtre, France (F.A., A.K., M.B., D.B., O.D.C.)
| | - Alain Fischer
- INSERM, UMR_S 1163, Laboratory of Normal and Pathological Homeostasis of the Immune System, Paris, France (M.K., I.M., A.F., G.d.S.B., G.M.).,Imagine Institute (M.K., I.M., A.F., G.d.S.B., G.M.).,Department of Immunology and Pediatric Hematology (A.F.)
| | - Delphine Borgel
- From the INSERM, UMR_S 1176, Paris-Sud University, Université Paris-Saclay, Le Kremlin-Bicêtre, France (F.A., A.K., M.B., D.B., O.D.C.).,Biological Hematology Service (D.B.), Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, France; and Collège de France, Paris (A.F.)
| | - Geneviève de Saint Basile
- INSERM, UMR_S 1163, Laboratory of Normal and Pathological Homeostasis of the Immune System, Paris, France (M.K., I.M., A.F., G.d.S.B., G.M.).,Imagine Institute (M.K., I.M., A.F., G.d.S.B., G.M.)
| | - Olivier D Christophe
- From the INSERM, UMR_S 1176, Paris-Sud University, Université Paris-Saclay, Le Kremlin-Bicêtre, France (F.A., A.K., M.B., D.B., O.D.C.)
| | - Gaël Ménasché
- INSERM, UMR_S 1163, Laboratory of Normal and Pathological Homeostasis of the Immune System, Paris, France (M.K., I.M., A.F., G.d.S.B., G.M.).,Imagine Institute (M.K., I.M., A.F., G.d.S.B., G.M.)
| |
Collapse
|
16
|
A novel mechanism regulating human platelet activation by MMP-2-mediated PAR1 biased signaling. Blood 2016; 129:883-895. [PMID: 28034890 DOI: 10.1182/blood-2016-06-724245] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 12/14/2016] [Indexed: 11/20/2022] Open
Abstract
Platelets contain and release several matrix metalloproteinases (MMPs). Among these, active MMP-2 enhances platelet aggregation by favoring the activation of phosphatidylinositol 3- kinase (PI3K) and contributes to arterial thrombosis. The platelet surface target of MMP-2 and the mechanism through which it primes platelets to respond to subsequent stimuli are still unknown. We show that active MMP-2 enhances platelet activation induced by weak stimuli by cleaving PAR1 at a noncanonical extracellular site different from the thrombin-cleavage site and thus initiates biased receptor signaling, triggering only some of the signaling pathways normally activated by full PAR1 agonism. The novel PAR1-tethered ligand exposed by MMP-2 stimulates PAR1-dependent Gq and G12/13 pathway activation, triggering p38-MAPK phosphorylation, Ca+2 fluxes, and PI3K activation, but not Gi signaling; this is insufficient to cause platelet aggregation, but it is enough to predispose platelets to fully respond to Gi-activating stimuli. Integrin αIIbβ3 is a necessary cofactor for PAR1 cleavage by MMP-2 by binding the MMP-2 hemopexin domain, thus favoring the interaction of the enzyme with PAR1. Our studies unravel a novel mechanism regulating platelet activation that involves the binding of MMP-2 to integrin αIIbβ3 and the subsequent cleavage of PAR1 by active MMP-2 at a noncanonical site, exposing a previously undescribed tethered ligand that triggers biased G-protein agonism and thus predisposes platelets to full activation by other stimuli. These results identify the MMP-2-αIIbβ3-PAR1 interaction as a potential target for the prevention of arterial thrombosis.
Collapse
|
17
|
Abstract
Protease signaling in cells elicits multiple physiologically important responses via protease-activated receptors (PARs). There are 4 members of this family of G-protein-coupled receptors (PAR1-4). PARs are activated by proteolysis of the N terminus to reveal a tethered ligand. The rate-limiting step of PAR signaling is determined by the efficiency of proteolysis of the N terminus, which is regulated by allosteric binding sites, cofactors, membrane localization, and receptor dimerization. This ultimately controls the initiation of PAR signaling. In addition, these factors also control the cellular response by directing signaling toward G-protein or β-arrestin pathways. PAR1 signaling on endothelial cells is controlled by the activating protease and heterodimerization with PAR2 or PAR3. As a consequence, the genetic and epigenetic control of PARs and their cofactors in physiologic and pathophysiologic conditions have the potential to influence cellular behavior. Recent studies have uncovered polymorphisms that result in PAR4 sequence variants with altered reactivity that interact to influence platelet response. This further demonstrates how interactions within the plasma membrane can control the physiological output. Understanding the structural rearrangement following PAR activation and how PARs are allosterically controlled within the plasma membrane will determine how best to target this family of receptors therapeutically. The purpose of this article is to review how signaling from PARs is influenced by alternative cleavage sites and the physical interactions within the membrane. Going forward, it will be important to relate the altered signaling to the molecular arrangement of PARs in the cell membrane and to determine how these may be influenced genetically.
Collapse
|
18
|
Thrombin-induced reactive oxygen species generation in platelets: A novel role for protease-activated receptor 4 and GPIbα. Redox Biol 2015; 6:640-647. [PMID: 26569550 PMCID: PMC4656914 DOI: 10.1016/j.redox.2015.10.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 10/23/2015] [Accepted: 10/23/2015] [Indexed: 11/24/2022] Open
Abstract
Background Platelets are essential for maintaining haemostasis and play a key role in the pathogenesis of cardiovascular disease. Upon ligation of platelet receptors through subendothelial matrix proteins, intracellular reactive oxygen species (ROS) are generated, further amplifying the platelet activation response. Thrombin, a potent platelet activator, can signal through GPIbα and protease-activated receptor (PAR) 1 and PAR4 on human platelets, and recently has been implicated in the generation of ROS. While ROS are known to have key roles in intra-platelet signalling and subsequent platelet activation, the precise receptors and signalling pathways involved in thrombin-induced ROS generation have yet to be fully elucidated. Objective To investigate the relative contribution of platelet GPIbα and PARs to thrombin-induced reactive oxygen species (ROS) generation. Methods and results Highly specific antagonists targeting PAR1 and PAR4, and the GPIbα-cleaving enzyme, Naja kaouthia (Nk) protease, were used in quantitative flow cytometry assays of thrombin-induced ROS production. Antagonists of PAR4 but not PAR1, inhibited thrombin-derived ROS generation. Removal of the GPIbα ligand binding region attenuated PAR4-induced and completely inhibited thrombin-induced ROS formation. Similarly, PAR4 deficiency in mice abolished thrombin-induced ROS generation. Additionally, GPIbα and PAR4-dependent ROS formation were shown to be mediated through focal adhesion kinase (FAK) and NADPH oxidase 1 (NOX1) proteins. Conclusions Both GPIbα and PAR4 are required for thrombin-induced ROS formation, suggesting a novel functional cooperation between GPIbα and PAR4. Our study identifies a novel role for PAR4 in mediating thrombin-induced ROS production that was not shared by PAR1. This suggests an independent signalling pathway in platelet activation that may be targeted therapeutically. PAR4 plays an important role in platelet-derived ROS generation. Thrombin-induced ROS generation in platelets require both GPIbα and PAR4. Potential functional association between GPIbα and PAR4 receptors and mouse and human platelets. GPIbα and PAR4-dependent ROS formation is mediated through FAK and NOX1 proteins.
Collapse
|
19
|
Duan K, Yu W, Li N. The Pathophysiology and Management of Acute Traumatic Coagulopathy. Clin Appl Thromb Hemost 2013; 21:645-52. [PMID: 24363215 DOI: 10.1177/1076029613516190] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Acute traumatic coagulopathy (ATC) is commonly seen among patients with severe injury and will lead to uncontrolled bleeding diathesis, which is an important contributor to trauma death. During the past 10 years, the understanding of the mechanism causing ATC has changed rapidly. The mechanisms for ATC are complicated. To date, the possible mechanisms include activation of protein C, shedding of endothelial glycocalyx, catecholamine release, platelet dysfunction, primary, and secondary fibrinolysis, with tissue injury and hypoperfusion as the triggers. Classic factors such as dilution, acidosis, and hypothermia can further aggravate the coagulopathy. Inflammation may have a potential effect on the onset and prognosis of ATC. With the aid of diagnostic device, the outcome can be improved through early and customized treatment. Antifibrinolytics such as tranexamic acid has some benefits in patients with bleeding trauma, especially in the early time. This review presents the current understanding of ATC mechanisms and management.
Collapse
Affiliation(s)
- Kaipeng Duan
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Wenkui Yu
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Ning Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| |
Collapse
|
20
|
Burzaco J, Conde M, Parada LA, Zugaza JL, Dehaye JP, Marino A. ATP antagonizes thrombin-induced signal transduction through 12(S)-HETE and cAMP. PLoS One 2013; 8:e67117. [PMID: 23826207 PMCID: PMC3691129 DOI: 10.1371/journal.pone.0067117] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 05/15/2013] [Indexed: 11/18/2022] Open
Abstract
In this study we have investigated the role of extracellular ATP on thrombin induced-platelet aggregation (TIPA) in washed human platelets. ATP inhibited TIPA in a dose-dependent manner and this inhibition was abolished by apyrase but not by adenosine deaminase (ADA) and it was reversed by extracellular magnesium. Antagonists of P2Y1 and P2Y12 receptors had no effect on this inhibition suggesting that a P2X receptor controlled ATP-mediated TIPA inhibition. ATP also blocked inositol phosphates (IP1, IP2, IP3) generation and [Ca(2+)]i mobilization induced by thrombin. Thrombin reduced cAMP levels which were restored in the presence of ATP. SQ-22536, an adenylate cyclase (AC) inhibitor, partially reduced the inhibition exerted by ATP on TIPA. 12-lipoxygenase (12-LO) inhibitors, nordihidroguaretic acid (NDGA) and 15(S)-hydroxy-5,8,11,13-eicosatetraenoic acid (15(S)-HETE), strongly prevented ATP-mediated TIPA inhibition. Additionally, ATP inhibited the increase of 12(S)-hydroxy-5,8,10,14-eicosatetraenoic acid (12(S)-HETE) induced by thrombin. Pretreatment with both SQ-22536 and NDGA almost completely abolished ATP-mediated TIPA inhibition. Our results describe for the first time that ATP implicates both AC and 12-LO pathways in the inhibition of human platelets aggregation in response to agonists.
Collapse
Affiliation(s)
- Jaione Burzaco
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country, Bilbao, Spain
| | - Manuel Conde
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country, Bilbao, Spain
| | - Luis A. Parada
- Instituto de Patología Experimental, Universidad Nacional de Salta, Salta, Argentina
| | - José L. Zugaza
- Department Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country, Bilbao, Spain
- Achucarro Basque Center for Neuroscience, Bizkaia Science and Technology Park, Zamudio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Jean-Paul Dehaye
- Biochemistry and Cellular Biology Laboratory, Institute of Pharmacy C.P. 205/3, Université Libre de Bruxelles, Brussels, Belgium
| | - Aida Marino
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country, Bilbao, Spain
| |
Collapse
|
21
|
Jiang L, Xu C, Yu S, Liu P, Luo D, Zhou Q, Gao C, Hu H. A critical role of thrombin/PAR-1 in ADP-induced platelet secretion and the second wave of aggregation. J Thromb Haemost 2013; 11:930-40. [PMID: 23406164 DOI: 10.1111/jth.12168] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 02/03/2013] [Indexed: 01/08/2023]
Abstract
BACKGROUND The stable or second wave of platelet aggregation often observed in ADP-stimulated platelet-rich plasma (PRP) with an artificially lowered extracellular calcium level has been attributed to enhanced thromboxane A2 (TXA2 ) generation and inhibition of ectonucleotidase activity. However, the role of thrombin in ADP-induced platelet secretion and the second wave of aggregation is unknown. OBJECTIVES AND METHODS We employed aggregometry, flow cytometry, immunoblotting and ELISA to determine whether and how thrombin participates in ADP-induced platelet secretion and the second wave of aggregation. RESULTS ADP induces a phosphoinositide 3-kinase (PI3K) pathway-dependent thrombin generation, presumably resulting from the cleavage of αII b β3 -associated prothrombin. Generated thrombin subsequently activates protease-activated receptor-1 (PAR-1) and mediates dense granule secretion and the second wave of platelet aggregation in ADP-stimulated citrated PRP. Thus, ADP-induced dense granule secretion and the second wave of platelet aggregation in PRP were similarly and non-additively blocked by thrombin inhibitor hirudin, PAR-1 antagonist SCH-79797 or PI3K inhibitor wortmannin. Moreover, ADP stimulation caused the dissociation of prothrombin from αII b β3 and an increased plasma thrombin level; both were prevented by wortmannin. Furthermore, the wortmannin-inhibited second wave of platelet aggregation by ADP was restored by a subaggregation concentration of PAR-1 activating peptide SFLLRN. Blocking TXA2 production with indomethacin or restoring extracellular calcium to physiological concentration did not influence this thrombin/PAR-1 dependence. CONCLUSIONS A PI3K-dependent thrombin generation and the resultant PAR-1 activation serve as an indispensable mechanism to relay the platelet activation process induced by ADP.
Collapse
Affiliation(s)
- L Jiang
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
A trauma patient's survival depends on the ability to control 2 opposing conditions, bleeding at the early phase and thrombosis at a late phase of trauma. The mixed existence of physiological responses for hemostasis and wound healing and pathological hemostatic responses makes it difficult to understand the mechanisms of the 2 stages of coagulopathy after trauma. Traumatic coagulopathy is multifactorial but disseminated intravascular coagulation (DIC) with the fibrinolytic phenotype is the predominant and initiative pathogenesis of coagulopathy at the early stage of trauma. High levels of inflammatory cytokines and severe tissue injuries activate the tissue-factor-dependent coagulation pathway followed by massive thrombin generation and its activation. Low levels of protein C and antithrombin induce insufficient coagulation control and the inhibition of the anticoagulation pathway. Primary and secondary fibrin(ogen)olysis is highly activated by the shock-induced tissue hypoxia and disseminated fibrin formation, respectively. Consumption coagulopathy and severe bleeding are subsequently observed in trauma patients. Persistently high levels of plasminogen activator inhibitor-1 expressed in the platelets and endothelium then change the DIC with the fibrinolytic phenotype into the thrombotic phenotype at approximately 24 to 48 hours after the onset of trauma. All of these changes coincide with the definition of DIC, which can be clearly distinguished from normal responses for hemostasis and wound healing by using sensitive molecular markers and DIC diagnostic criteria such as those outlined by the Japanese Association for Acute Medicine and the International Society on Thrombosis and Haemostasis. Treatments of DIC with the fibrinolytic phenotype involve the surgical repair of the trauma, improvement of shock, and the rapid and sufficient replacement of platelet concentrate, fresh frozen plasma, and depleted coagulation factors. The administration of an antifibrinolytic agent (tranexamic acid) may reduce the risk of death in bleeding trauma patients associated with this type of DIC.
Collapse
|
23
|
Harper MT, Poole AW. PKC inhibition markedly enhances Ca2+ signaling and phosphatidylserine exposure downstream of protease-activated receptor-1 but not protease-activated receptor-4 in human platelets. J Thromb Haemost 2011; 9:1599-607. [PMID: 21649850 DOI: 10.1111/j.1538-7836.2011.04393.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Cytosolic calcium concentration is a critical regulator of platelet activation, and so platelet Ca(2+) signaling must be tightly controlled. Thrombin-induced Ca(2+) signaling is enhanced by inhibitors of protein kinase C (PKC), suggesting that PKC negatively regulates the Ca(2+) signal, although the mechanisms by which this occurs and its physiological relevance are still unclear. OBJECTIVES To investigate the mechanisms by which PKC inhibitors enhance thrombin-induced Ca(2+) signaling, and to determine the importance of this pathway in platelet activation. METHODS Cytosolic Ca(2+) signaling was monitored in fura-2-loaded human platelets. Phosphatidylserine (PS) exposure, a marker of platelet procoagulant activity, was measured by annexin V binding and flow cytometry. RESULTS PKC inhibition by bisindolylmaleimide-I (BIM-I) enhanced α-thrombin-induced Ca(2+) signaling in a concentration-dependent manner. PAR1 signaling, activated by SFLLRN, was enhanced much more strongly than PAR4, activated by AYPGKF or γ-thrombin, which is a potent PAR4 agonist but a poor activator of PAR1. BIM-I had little effect on α-thrombin-induced signaling following treatment with the PAR1 antagonist, SCH-79797. BIM-I enhanced Ca(2+) release from intracellular stores and Ca(2+) entry, as assessed by Mn(2+) quench. However, the plasma membrane Ca(2+) ATPase inhibitor, 5(6)-carboxyeosin, did not prevent the effect of BIM-I. PKC inhibition strongly enhanced α-thrombin-induced PS exposure, which was reversed by blockade of PAR1. CONCLUSIONS Together, these data show that when PAR1 is stimulated, PKC negatively regulates Ca(2+) release and Ca(2+) entry, which leads to reduced platelet PS exposure.
Collapse
Affiliation(s)
- M T Harper
- School of Physiology and Pharmacology, University of Bristol, Bristol, UK.
| | | |
Collapse
|
24
|
Harper MT, Sage SO. Actin polymerisation regulates thrombin-evoked Ca2+signalling after activation of PAR-4 but not PAR-1 in human platelets. Platelets 2009; 17:134-42. [PMID: 16702038 DOI: 10.1080/09537100500441218] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The role of actin polymerisation in regulating thrombin-evoked Ca(2+) signalling was investigated in human platelets. We have previously reported that cytochalasin D (Cyt D) inhibits thapsigargin-evoked store-operated Ca(2+) entry (SOCE), which is believed to contribute a major component of thrombin-evoked Ca(2+) entry in platelets. In contrast, Cyt D increased thrombin-evoked Ca(2+) entry to 147.5 +/- 9.2% and Sr(2+) entry to 134.2 +/- 6.4% of control. Similar results were obtained with latrunculin A. This potentiation was not affected if protein kinase C was inhibited using Ro-31-8220, suggesting that it did not involve PKC-dependent non-capacitative Ca(2+) entry. Ca(2+) entry evoked by the PAR-4 agonist, AYPGKF, was increased to 133.7 +/- 12.8% of control by Cyt D, whereas Ca(2+) signalling evoked by the PAR-1 agonist, SFLLRN, was unaffected. The PAR-4 antagonist, tcY-NH(2), abolished the effect of Cyt D on thrombin-evoked Ca(2+) entry. Biotinylation of cell-surface proteins showed that PAR-4 was internalised after stimulation by thrombin. Cyt D reduced this internalisation. These data suggest that Cyt D prevents the internalisation of PAR-4, which may lead to prolonged signalling from this receptor. This may mask a direct effect of Cyt D on the activation of SOCE after the activation of PAR-4.
Collapse
Affiliation(s)
- Matthew T Harper
- Department of Physiology, University of Cambridge, Downing Street, Cambridge, UK.
| | | |
Collapse
|
25
|
|
26
|
Pabón D, Jayo A, Xie J, Lastres P, González-Manchón C. Thrombin induces GPIb-IX-mediated fibrin binding to alphaIIbbeta3 in a reconstituted Chinese hamster ovary cell model. J Thromb Haemost 2006; 4:2238-47. [PMID: 16999852 DOI: 10.1111/j.1538-7836.2006.02141.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The interaction of thrombin with platelet glycoprotein (GP) Ib-IX-V has been recently suggested to induce fibrin-dependent platelet aggregation associated with signaling events. The approaches used to avoid the protease-activated receptor (PAR) thrombin receptors in platelets have provided controversial conclusions regarding the precise mechanism and molecules involved in the response. OBJECTIVES In the present study, we developed a cellular model to investigate the functional consequences following the binding of thrombin to GPIb-IX. METHODS We used Chinese hamster ovary (CHO) cells stably expressing human alpha(IIb)beta(3) and/or GPIb-IX complexes (CHO-alpha(IIb)beta(3)-IbIX cells) to analyze the effect of thrombin on the binding of polymerizing fibrin by using fluorescein isothiocyanate-fibrinogen as precursor. RESULTS Thrombin induces, in a dose-dependent manner, the binding of polymerizing fibrin to CHO-alpha(IIb)beta(3)-IbIX cells. This response is not observed in cells expressing only one of the receptors, and it can be blocked by monoclonal antibodies against alpha(IIb)beta(3) and GPIbalpha. We show that the reaction is not due to simple cell trapping by the fibrin clot, and provide data supporting a role of a signaling pathway in which the 14-3-3zeta adaptor and calcium-calmodulin-dependent events are involved. CONCLUSIONS The present data support a significant role of GPIb-IX and alpha(IIb)beta(3) receptors in an alternative fibrin-mediated pathway of platelet activation induced by thrombin.
Collapse
Affiliation(s)
- D Pabón
- Department of Molecular and Cellular Pathophysiology, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | | | | | | | | |
Collapse
|
27
|
Holinstat M, Voss B, Bilodeau ML, McLaughlin JN, Cleator J, Hamm HE. PAR4, but not PAR1, signals human platelet aggregation via Ca2+ mobilization and synergistic P2Y12 receptor activation. J Biol Chem 2006; 281:26665-74. [PMID: 16837456 PMCID: PMC3035573 DOI: 10.1074/jbc.m602174200] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Regulation of platelet activation plays a central role in hemostasis and pathophysiological processes such as coronary artery disease. Thrombin is the most potent activator of platelets. Human platelets express two thrombin receptors, PAR1 and PAR4, both of which signal platelet activation. Evidence is lacking on the mechanism by which PAR1 and PAR4 may differentially signal platelet aggregation. Here we show that at the relatively high concentration of agonist most likely found at the site of a local thrombus, dual inhibition of the P2Y12 receptor and calcium mobilization result in a complete inhibition of PAR4-induced aggregation, while having no effect on either thrombin or PAR1-mediated platelet aggregation. Both PAR1- and PAR4mediated aggregation are independent of calcium mobilization. Furthermore, we show that P2Y12 receptor activation is not required for protease-activated receptor-mediated aggregation at higher agonist concentrations and is only partially required for Rap1 as well as GPIIbIIIa activation. P2Y12 receptor inhibitors clinically in use such as clopidogrel are postulated to decrease platelet aggregation through partial inhibition of PAR1 signaling. Our data, however, indicate that at high local concentrations of thrombin, it is the signaling through PAR4 rather than PAR1 that may be regulated through purinergic feedback. Thus, our data identify an intra-platelet mechanism that may function as a future site for therapeutic intervention.
Collapse
Affiliation(s)
- Michael Holinstat
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6600
| | - Bryan Voss
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6600
| | - Matthew L. Bilodeau
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6600
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6600
| | | | - John Cleator
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6600
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6600
| | - Heidi E. Hamm
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6600
| |
Collapse
|
28
|
Wu CC, Teng CM. Comparison of the effects of PAR1 antagonists, PAR4 antagonists, and their combinations on thrombin-induced human platelet activation. Eur J Pharmacol 2006; 546:142-7. [PMID: 16890935 DOI: 10.1016/j.ejphar.2006.07.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2005] [Revised: 05/26/2006] [Accepted: 07/07/2006] [Indexed: 10/24/2022]
Abstract
Thrombin activates human platelets through proteolytic activation of two protease-activated receptors (PARs), PAR1 and PAR4. In the present study, we show that, RWJ-56110, a potent synthetic PAR1 antagonist, inhibited platelet aggregation caused by a low concentration (0.05 U/ml) of thrombin, but lost its effectiveness when higher concentrations of thrombin were used as stimulators. YD-3, a non-peptide PAR4 antagonist, alone had little or no effect on thrombin-induced platelet aggregation, significantly enhanced the anti-aggregatory activity of PAR1 antagonist. In addition, we demonstrate for the first time that P-selectin expression in thrombin-stimulated platelets can be synergistically prevented by combined treatment of PAR1 antagonist and PAR4 antagonist. These results indicate that thrombin-induced platelet activation cannot be effectively inhibited by just blocking either single thrombin receptor pathway, and suggest a rationale for potential combination therapy in arterial thrombosis.
Collapse
Affiliation(s)
- Chin-Chung Wu
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | | |
Collapse
|
29
|
Tantry US, Bliden KP, Gurbel PA. Resistance to antiplatelet drugs: current status and future research. Expert Opin Pharmacother 2005; 6:2027-45. [PMID: 16197357 DOI: 10.1517/14656566.6.12.2027] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Platelet reactivity and activation are important factors during the development of atherothrombotic processes and subsequent ischaemic complications. Pharmacological agents that suppress platelet function are proved to be the most efficient in the prevention and treatment of thrombotic complications. As the activation of platelets during thrombus generation involves many complex and redundant pathways, simultaneous use of different antiplatelet drugs that are directed against different targets have been effective in reducing adverse clinical events. The main antiplatelet drugs are aspirin (which inhibits thromboxane synthesis), thienopyridines (which block P2Y12 receptors) and glycoprotein IIb/IIIa antagonists (which block glycoprotein IIb/IIIa receptors). In recent years, resistance or nonresponsiveness to antiplatelet therapy has been reported and, more importantly, are linked to the occurrence of adverse cardiovascular events. New treatment strategies to overcome nonresponsiveness are being sought. A focus on the development of simple, reproducible and user friendly point-of-care methods to determine aspirin/clopidogrel responsiveness should be undertaken to assist clinicians in tailoring antiplatelet therapy to the individual patient.
Collapse
Affiliation(s)
- Udaya S Tantry
- Sinai Center for Thrombosis Research, Hoffberger Building, Suite 56, 2401 W. Belvedere Ave, Baltimore, MD 21215, USA
| | | | | |
Collapse
|
30
|
Xu CS, Han HP, Yuan JY, Chang CF, Li WQ, Yang KJ, Zhao LF, Li YC, Zhang HY, Salman R, Zhang JB. Gene expression difference in regenerating rat liver after 0-36-40-44h short interval successive partial hepatectomy. Shijie Huaren Xiaohua Zazhi 2004; 12:654-663. [DOI: 10.11569/wcjd.v12.i3.654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To identify genes related to rat liver regeneration (LR) after 0-36-40-44h short interval successive partial hepatectomy (SISPH) and to analyze their action and expression profile in LR.
METHODS: A cDNA microarray containing 551 elements (liver chip) was made to analyze extensively expression changes of them in 0-36-40-44h SISPH, which were selected from subtractive cDNA libraries of the LR. Cluster analysis of these gene expression profile was performed by Genemath.
RESULTS: Among the selected 551 cDNA, 157 were up- ordown-regulated more than twofold at one or more time points. Of the 157 elements, 86 were up-regulated and 71 down-regulated, and 70 were not reported and 87 were reported, which had not been previously reported to be involved in LR. By cluster analysis and generalization analysis, 6 distinct temporal induction or suppression patterns showed that immediate induction, intermediate induction, late induction, immediate suppression, intermediate suppression, and late suppression. Comparison of the gene expression in SISPH with after PH found that 38 genes were specially altered in SISPH, and the expression trends for other 119 genes were similar between SISPH and PH, except of the various abundance at the different time points.
CONCLUSION: In 0-36-40-44h SISPH, the numbers of the up-regulated and down-regulated genes show no apparent difference. The genes expressed lately are more than that immediately, and much more than that intermediately. The genes expressed abundantly are much less than that increased 2-5 folds.
Collapse
|
31
|
Adam F, Bouton MC, Huisse MG, Jandrot-Perrus M. Thrombin interaction with platelet membrane glycoprotein Ibα. Trends Mol Med 2003; 9:461-4. [PMID: 14604821 DOI: 10.1016/j.molmed.2003.09.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The interaction of thrombin with platelet glycoprotein Ibalpha (GPIb alpha) is required for optimal platelet activation. The crystal structures of platelet GPIb alpha bound to thrombin reported by Dumas et al. and Celikel et al. both reveal the simultaneous interaction of GPIb alpha with thrombin exosites I and II but differ markedly regarding how the two proteins interact. The possible consequences on thrombus formation of thrombin interacting with GPIb alpha are discussed in light of these new data.
Collapse
Affiliation(s)
- Frédéric Adam
- INSERM E348, Faculté de Médecine Xavier Bichat, 16 rue Henri Huchard, 75870 Paris Cedex 18, France
| | | | | | | |
Collapse
|