51
|
Kato Y, Nishida O, Kuriyama N, Nakamura T, Kawaji T, Onouchi T, Hasegawa D, Shimomura Y. Effects of Thrombomodulin in Reducing Lethality and Suppressing Neutrophil Extracellular Trap Formation in the Lungs and Liver in a Lipopolysaccharide-Induced Murine Septic Shock Model. Int J Mol Sci 2021; 22:4933. [PMID: 34066510 PMCID: PMC8124404 DOI: 10.3390/ijms22094933] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 12/19/2022] Open
Abstract
Neutrophil extracellular trap (NET) formation, an innate immune system response, is associated with thrombogenesis and vascular endothelial injury. Circulatory disorders due to microvascular thrombogenesis are one of the principal causes of organ damage. NET formation in organs contributes to the exacerbation of sepsis, which is defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection. We have previously reported that recombinant human soluble thrombomodulin (rTM) reduces lipopolysaccharide (LPS)-induced NET formation in vitro. Here, we aimed to show that thrombomodulin (TM)-mediated suppression of NET formation protects against organ damage in sepsis. Mice were injected intraperitoneally (i.p.) with 10 mg/kg LPS. rTM (6 mg/kg/day) or saline was administered i.p. 1 h after LPS injection. In the LPS-induced murine septic shock model, extracellular histones, which are components of NETs, were observed in the liver and lungs. In addition, the serum cytokine (interleukin-1β (IL-1β), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), macrophage chemotactic protein-1 (MCP-1), and interleukin-10 (IL-10)) levels were increased. The administration of rTM in this model prevented NET formation in the organs and suppressed the increase in the levels of all cytokines except IL-1β. Furthermore, the survival rate improved. We provide a novel role of TM in treating inflammation and NETs in organs during sepsis.
Collapse
Affiliation(s)
- Yu Kato
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake 470-1192, Japan; (Y.K.); (O.N.); (N.K.); (T.N.); (T.K.); (D.H.)
| | - Osamu Nishida
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake 470-1192, Japan; (Y.K.); (O.N.); (N.K.); (T.N.); (T.K.); (D.H.)
| | - Naohide Kuriyama
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake 470-1192, Japan; (Y.K.); (O.N.); (N.K.); (T.N.); (T.K.); (D.H.)
| | - Tomoyuki Nakamura
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake 470-1192, Japan; (Y.K.); (O.N.); (N.K.); (T.N.); (T.K.); (D.H.)
| | - Takahiro Kawaji
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake 470-1192, Japan; (Y.K.); (O.N.); (N.K.); (T.N.); (T.K.); (D.H.)
| | - Takanori Onouchi
- Center for Joint Research Facilities Support, Research Promotion and Support Headquarters, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake 470-1192, Japan;
| | - Daisuke Hasegawa
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake 470-1192, Japan; (Y.K.); (O.N.); (N.K.); (T.N.); (T.K.); (D.H.)
| | - Yasuyo Shimomura
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake 470-1192, Japan; (Y.K.); (O.N.); (N.K.); (T.N.); (T.K.); (D.H.)
| |
Collapse
|
52
|
Shevchuk O, Begonja AJ, Gambaryan S, Totzeck M, Rassaf T, Huber TB, Greinacher A, Renne T, Sickmann A. Proteomics: A Tool to Study Platelet Function. Int J Mol Sci 2021; 22:ijms22094776. [PMID: 33946341 PMCID: PMC8125008 DOI: 10.3390/ijms22094776] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 12/22/2022] Open
Abstract
Platelets are components of the blood that are highly reactive, and they quickly respond to multiple physiological and pathophysiological processes. In the last decade, it became clear that platelets are the key components of circulation, linking hemostasis, innate, and acquired immunity. Protein composition, localization, and activity are crucial for platelet function and regulation. The current state of mass spectrometry-based proteomics has tremendous potential to identify and quantify thousands of proteins from a minimal amount of material, unravel multiple post-translational modifications, and monitor platelet activity during drug treatments. This review focuses on the role of proteomics in understanding the molecular basics of the classical and newly emerging functions of platelets. including the recently described role of platelets in immunology and the development of COVID-19.The state-of-the-art proteomic technologies and their application in studying platelet biogenesis, signaling, and storage are described, and the potential of newly appeared trapped ion mobility spectrometry (TIMS) is highlighted. Additionally, implementing proteomic methods in platelet transfusion medicine, and as a diagnostic and prognostic tool, is discussed.
Collapse
Affiliation(s)
- Olga Shevchuk
- Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V, Bunsen-Kirchhoff-Straße 11, 44139 Dortmund, Germany
- Department of Immunodynamics, Institute of Experimental Immunology and Imaging, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
- Correspondence: (O.S.); (A.S.)
| | - Antonija Jurak Begonja
- Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia;
| | - Stepan Gambaryan
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Torez pr. 44, 194223 St. Petersburg, Russia;
| | - Matthias Totzeck
- West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany; (M.T.); (T.R.)
| | - Tienush Rassaf
- West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany; (M.T.); (T.R.)
| | - Tobias B. Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Andreas Greinacher
- Institut für Immunologie und Transfusionsmedizin, Universitätsmedizin Greifswald, Sauerbruchstraße, 17475 Greifswald, Germany;
| | - Thomas Renne
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany;
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V, Bunsen-Kirchhoff-Straße 11, 44139 Dortmund, Germany
- Medizinisches Proteom-Center (MPC), Medizinische Fakultät, Ruhr-Universität Bochum, 44801 Bochum, Germany
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen AB24 3FX, UK
- Correspondence: (O.S.); (A.S.)
| |
Collapse
|
53
|
Mariscal A, Zamora C, Magallares B, Salman-Monte TC, Ortiz MÀ, Díaz-Torné C, Castellví I, Corominas H, Vidal S. Phenotypic and Functional Consequences of PLT Binding to Monocytes and Its Association with Clinical Features in SLE. Int J Mol Sci 2021; 22:4719. [PMID: 33947017 PMCID: PMC8125177 DOI: 10.3390/ijms22094719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022] Open
Abstract
Platelets (PLTs) can modulate the immune system through the release of soluble mediators or through interaction with immune cells. Monocytes are the main immune cells that bind with PLTs, and this interaction is increased in several inflammatory and autoimmune conditions, including systemic lupus erythematosus (SLE). Our aim was to characterize the phenotypic and functional consequences of PLT binding to monocytes in healthy donors (HD) and in SLE and to relate it to the pathogenesis of SLE. We analyzed the phenotypic and functional features of monocytes with non-activated and activated bound PLTs by flow cytometry. We observed that monocytes with bound PLTs and especially those with activated PLTs have an up-regulated HLA-DR, CD86, CD54, CD16 and CD64 expression. Monocytes with bound PLTs also have an increased capacity for phagocytosis, though not for efferocytosis. In addition, monocytes with bound PLTs have increased IL-10, but not TNF-α, secretion. The altered phenotypic and functional features are comparable in SLE and HD monocytes and in bound PLTs. However, the percentages of monocytes with bound PLTs are significantly higher in SLE patients and are associated with undetectable levels of anti-dsDNA antibodies and hematuria, and with normal C3 and albumin/creatinine levels. Our results suggest that PLTs have a modulatory influence on monocytes and that this effect may be highlighted by an increased binding of PLTs to monocytes in autoimmune conditions.
Collapse
Affiliation(s)
- Anaís Mariscal
- Immunology Department, Hospital de la Santa Creu I Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain;
| | - Carlos Zamora
- Laboratory of Inflammatory Diseases, Hospital de la Santa Creu I Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (C.Z.); (M.À.O.)
| | - Berta Magallares
- Rheumatology Department, Hospital de la Santa Creu I Sant Pau, 08041 Barcelona, Spain; (B.M.); (C.D.-T.); (I.C.); (H.C.)
| | | | - Mª Àngels Ortiz
- Laboratory of Inflammatory Diseases, Hospital de la Santa Creu I Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (C.Z.); (M.À.O.)
| | - Cesar Díaz-Torné
- Rheumatology Department, Hospital de la Santa Creu I Sant Pau, 08041 Barcelona, Spain; (B.M.); (C.D.-T.); (I.C.); (H.C.)
| | - Iván Castellví
- Rheumatology Department, Hospital de la Santa Creu I Sant Pau, 08041 Barcelona, Spain; (B.M.); (C.D.-T.); (I.C.); (H.C.)
| | - Héctor Corominas
- Rheumatology Department, Hospital de la Santa Creu I Sant Pau, 08041 Barcelona, Spain; (B.M.); (C.D.-T.); (I.C.); (H.C.)
| | - Silvia Vidal
- Laboratory of Inflammatory Diseases, Hospital de la Santa Creu I Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (C.Z.); (M.À.O.)
| |
Collapse
|
54
|
Pentraxin 3 expression in lungs and neutrophils of calves. Vet Immunol Immunopathol 2021; 236:110251. [PMID: 33901710 DOI: 10.1016/j.vetimm.2021.110251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/15/2021] [Accepted: 04/18/2021] [Indexed: 11/23/2022]
Abstract
Bacterial lung disease caused by Mannheimia haemolytica inflict significant mortality and morbidity resulting in enormous economic losses to cattle industry. The use of antibiotics is becoming more challenging because of development of anti-microbial resistance. The innate immune system plays a critical role in the initiation of immune response in the lung. Pentraxin 3 (PTX3), a pattern-recognition receptor is produced at sites of inflammation by many cell types, recognizes and binds to many pathogens, activates the complement cascade, and has a role in the clearance of apoptotic and necrotic cells. Because there are very few data on the expression of PTX3 in the lungs, we examined PTX3 expression in lungs of normal and M. haemolytica-infected calves and normal and E. coli lipopolysaccharide-treated cattle neutrophils using light and electron microscopic immunochemistry and Western blots. Immunohistology showed the presence of PTX3 in airway epithelial cells, alveolar septa and macrophages in normal and inflamed lungs of calves and the blots showed a significant increase in the expression of PTX3 in lungs from infected calves. Immuno-gold electron microscopy showed PTX3 in the nuclei, cytoplasm, and vesicular organelles of alveolar macrophages, endothelial cells and pulmonary intravascular macrophages (PIMs). Immunohistochemical staining for PTX3 in peripheral blood neutrophils shows an altered staining pattern in neutrophils stimulated with lipopolysachharide (LPS). However, western blots no significant change in PTX3 amount in LPS-treated neutrophils compared to the controls. These are the first data on the expression of PTX3 in the lungs and the neutrophils of cattle which may add to our understanding of innate immunity in cattle lungs.
Collapse
|
55
|
Allaoui A, Khawaja AA, Badad O, Naciri M, Lordkipanidzé M, Guessous F, Zaid Y. Platelet Function in Viral Immunity and SARS-CoV-2 Infection. Semin Thromb Hemost 2021; 47:419-426. [PMID: 33851385 DOI: 10.1055/s-0041-1726033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Platelets, as nonnucleated blood components, are classically recognized for their pivotal role in hemostasis. In recent years, however, accumulating evidence points to a nonhemostatic role for platelets, as active participants in the inflammatory and immune responses to microbial organisms in infectious diseases. This stems from the ability of activated platelets to secrete a plethora of immunomodulatory cytokines and chemokines, as well as directly interplaying with viral receptors. While much attention has been given to the role of the cytokine storm in the severity of the coronavirus disease 2019 (COVID-19), less is known about the contribution of platelets to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Here, we give a brief overview on the platelet contribution to antiviral immunity and response during SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Afaf Allaoui
- Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Akif A Khawaja
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Oussama Badad
- Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco.,Department of Plant, Southern Illinois University, Carbondale, Illinois
| | - Mariam Naciri
- Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Marie Lordkipanidzé
- Research Center, Montreal Heart Institute, Montréal, Quebec, Canada.,Faculty of pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Fadila Guessous
- Microbiology, Immunology and Cancer Biology, School of Medicine, University of Virginia, Charlottesville, Virginia.,Department of Biological Sciences, Faculty of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Younes Zaid
- Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco.,Research Center of Abulcasis University of Health Sciences, Cheikh Zaïd Hospital, Rabat, Morocco
| |
Collapse
|
56
|
El-Kadiry AEH, Merhi Y. The Role of the Proteasome in Platelet Function. Int J Mol Sci 2021; 22:3999. [PMID: 33924425 PMCID: PMC8069084 DOI: 10.3390/ijms22083999] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023] Open
Abstract
Platelets are megakaryocyte-derived acellular fragments prepped to maintain primary hemostasis and thrombosis by preserving vascular integrity. Although they lack nuclei, platelets harbor functional genomic mediators that bolster platelet activity in a signal-specific manner by performing limited de novo protein synthesis. Furthermore, despite their limited protein synthesis, platelets are equipped with multiple protein degradation mechanisms, such as the proteasome. In nucleated cells, the functions of the proteasome are well established and primarily include proteostasis among a myriad of other signaling processes. However, the role of proteasome-mediated protein degradation in platelets remains elusive. In this review article, we recapitulate the developing literature on the functions of the proteasome in platelets, discussing its emerging regulatory role in platelet viability and function and highlighting how its functional coupling with the transcription factor NF-κB constitutes a novel potential therapeutic target in atherothrombotic diseases.
Collapse
Affiliation(s)
- Abed El-Hakim El-Kadiry
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Research Centre, Montreal, QC H1T 1C8, Canada;
- Biomedical Sciences Program, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Yahye Merhi
- Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Research Centre, Montreal, QC H1T 1C8, Canada;
- Biomedical Sciences Program, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
57
|
Corcione N, Romano S, Morello A, Ferraro P, Cimmino M, Albanese M, Tufano M, Capasso D, Buonpane S, Giordano S, Pepe M, Biondi-Zoccai G, Romano MF, Giordano A. Thrombocytopenia Complicating Transcatheter Aortic Valve Implantation: Differences Between Two New-Generation Devices. J Cardiovasc Transl Res 2021; 14:1104-1113. [PMID: 33721196 PMCID: PMC8651580 DOI: 10.1007/s12265-021-10117-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 02/25/2021] [Indexed: 02/08/2023]
Abstract
Thrombocytopenia after TAVI is common and clinically detrimental. Retrospectively, we observed Portico recipients had a more profound platelet drop than Evolut recipients. We thus investigated periprocedural platelet damage and/orpro-inflammatory state in 64 TAVI recipients at baseline and after implantation. Platelet damage was assessed by annexin V staining and monocyte-phagocytic phenotype was assessed according to CD14/CD36 expression. Serum cytokines were measured in 20 patients. The formaldehyde-based storage solution altered platelets. When, before being loaded onto the delivery system, Portico underwent one additional flushing to those recommended, the receiving patients showed thrombocytopenia, platelet damage, and CD36-monocyte count were mitigated. A general increase in IL-6 was recorded in overall TAVI recipients, but a high serum level of IL-8, a potent thrombocytopenia inducer, was measured in Portico recipients only, including those with extra-rinsed valve. Our study suggests a platelet-injury effect by storage-solution and generates the hypothesis of a role for the biomaterial in stimulating innate-immunity. Larger prospective studies are needed. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Nicola Corcione
- Unità Operativa di Interventistica Cardiovascolare, Pineta Grande Hospital, Castel Volturno, Italy
| | - Simona Romano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Pansini, 5, 80131, Napoli, Italy
| | - Alberto Morello
- Unità Operativa di Interventistica Cardiovascolare, Pineta Grande Hospital, Castel Volturno, Italy
| | - Paolo Ferraro
- Unità Operativa di Emodinamica, Santa Lucia Hospital, San Giuseppe Vesuviano, Italy
| | - Michele Cimmino
- Unità Operativa di Emodinamica, Santa Lucia Hospital, San Giuseppe Vesuviano, Italy
| | - Michele Albanese
- Unità Operativa di Emodinamica, Santa Lucia Hospital, San Giuseppe Vesuviano, Italy
| | - Martina Tufano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Pansini, 5, 80131, Napoli, Italy
| | - Daniela Capasso
- Dipartimento Cuore U.O.C. Cardiologia-UTIC, Pineta Grande Hospital, Castel Volturno, Italy
| | - Salvatore Buonpane
- Unità di Medicina di Laboratorio, Pineta Grande Hospital, Castel Volturno, Italy
| | - Salvatore Giordano
- Unità Operativa di Interventistica Cardiovascolare, Pineta Grande Hospital, Castel Volturno, Italy
| | - Martino Pepe
- Division of Cardiology, Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,Mediterranea Cardiocentro, Napoli, Italy
| | - Maria Fiammetta Romano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Pansini, 5, 80131, Napoli, Italy.
| | - Arturo Giordano
- Unità Operativa di Interventistica Cardiovascolare, Pineta Grande Hospital, Castel Volturno, Italy
| |
Collapse
|
58
|
Cognasse F, Hally K, Fauteux-Daniel S, Eyraud MA, Arthaud CA, Fagan J, Mismetti P, Hamzeh-Cognasse H, Laradi S, Garraud O, Larsen P. Effects and Side Effects of Platelet Transfusion. Hamostaseologie 2021; 41:128-135. [PMID: 33711849 DOI: 10.1055/a-1347-6551] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aside from their canonical role in hemostasis, it is increasingly recognized that platelets have inflammatory functions and can regulate both adaptive and innate immune responses. The main topic this review aims to cover is the proinflammatory effects and side effects of platelet transfusion. Platelets prepared for transfusion are subject to stress injury upon collection, preparation, and storage. With these types of stress, they undergo morphologic, metabolic, and functional modulations which are likely to induce platelet activation and the release of biological response modifiers (BRMs). As a consequence, platelet concentrates (PCs) accumulate BRMs during processing and storage, and these BRMs are ultimately transfused alongside platelets. It has been shown that BRMs present in PCs can induce immune responses and posttransfusion reactions in the transfusion recipient. Several recent reports within the transfusion literature have investigated the concept of platelets as immune cells. Nevertheless, current and future investigations will face the challenge of encompassing the immunological role of platelets in the scope of transfusion.
Collapse
Affiliation(s)
- Fabrice Cognasse
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France.,SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Kathryn Hally
- Department of Surgery and Anaesthesia, University of Otago, Wellington, New Zealand.,Wellington Cardiovascular Research Group, Wellington, New Zealand.,School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Sebastien Fauteux-Daniel
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France.,SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Marie-Ange Eyraud
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France.,SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Charles-Antoine Arthaud
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France.,SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Jocelyne Fagan
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France.,SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Patrick Mismetti
- SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Hind Hamzeh-Cognasse
- SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Sandrine Laradi
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France.,SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Olivier Garraud
- SAINBIOSE, INSERM U1059, University of Lyon, Université Jean-Monnet-Saint-Etienne, France, France
| | - Peter Larsen
- Department of Surgery and Anaesthesia, University of Otago, Wellington, New Zealand.,Wellington Cardiovascular Research Group, Wellington, New Zealand.,School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| |
Collapse
|
59
|
Fu G, Deng M, Neal MD, Billiar TR, Scott MJ. Platelet-Monocyte Aggregates: Understanding Mechanisms and Functions in Sepsis. Shock 2021; 55:156-166. [PMID: 32694394 PMCID: PMC8008955 DOI: 10.1097/shk.0000000000001619] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
ABSTRACT Platelets have been shown to play an important immunomodulatory role in the pathogenesis of various diseases through their interactions with other immune and nonimmune cells. Sepsis is a major cause of death in the United States, and many of the mechanisms driving sepsis pathology are still unresolved. Monocytes have recently received increasing attention in sepsis pathogenesis, and multiple studies have associated increased levels of platelet-monocyte aggregates observed early in sepsis with clinical outcomes in sepsis patients. These findings suggest platelet-monocyte aggregates may be an important prognostic indicator. However, the mechanisms leading to platelet interaction and aggregation with monocytes, and the effects of aggregation during sepsis are still poorly defined. There are few studies that have really investigated functions of platelets and monocytes together, despite a large body of research showing separate functions of platelets and monocytes in inflammation and immune responses during sepsis. The goal of this review is to provide insights into what we do know about mechanisms and biological meanings of platelet-monocyte interactions, as well as some of the technical challenges and limitations involved in studying this important potential mechanism in sepsis pathogenesis. Improving our understanding of platelet and monocyte biology in sepsis may result in identification of novel targets that can be used to positively affect outcomes in sepsis.
Collapse
Affiliation(s)
- Guang Fu
- Department of General Surgery, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China (visiting scholar in Pittsburgh 2018-09/2020-09)
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Meihong Deng
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Matthew D. Neal
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Trauma Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Trauma Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Melanie J. Scott
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Trauma Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
60
|
The necroptotic cell death pathway operates in megakaryocytes, but not in platelet synthesis. Cell Death Dis 2021; 12:133. [PMID: 33510145 PMCID: PMC7843594 DOI: 10.1038/s41419-021-03418-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 02/06/2023]
Abstract
Necroptosis is a pro-inflammatory cell death program executed by the terminal effector, mixed lineage kinase domain-like (MLKL). Previous studies suggested a role for the necroptotic machinery in platelets, where loss of MLKL or its upstream regulator, RIPK3 kinase, impacted thrombosis and haemostasis. However, it remains unknown whether necroptosis operates within megakaryocytes, the progenitors of platelets, and whether necroptotic cell death might contribute to or diminish platelet production. Here, we demonstrate that megakaryocytes possess a functional necroptosis signalling cascade. Necroptosis activation leads to phosphorylation of MLKL, loss of viability and cell swelling. Analyses at steady state and post antibody-mediated thrombocytopenia revealed that platelet production was normal in the absence of MLKL, however, platelet activation and haemostasis were impaired with prolonged tail re-bleeding times. We conclude that MLKL plays a role in regulating platelet function and haemostasis and that necroptosis signalling in megakaryocytes is dispensable for platelet production.
Collapse
|
61
|
Coenen DM, Heinzmann ACA, Karel MFA, Cosemans JMEM, Koenen RR. The multifaceted contribution of platelets in the emergence and aftermath of acute cardiovascular events. Atherosclerosis 2021; 319:132-141. [PMID: 33468314 DOI: 10.1016/j.atherosclerosis.2020.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/17/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022]
Abstract
Atherosclerosis is an underlying cause of a broad array of cardiovascular diseases characterized by plaques, arterial wall thickening initiated by hyperlipidemia, pro-inflammatory signals, endothelial dysfunction and the influx of inflammatory cells. By still incompletely characterized mechanisms, these plaques can destabilize or erode, leading to thrombosis and blood vessel occlusion and becomes clinically manifest as angina pectoris, myocardial infarction (MI) or stroke. Among the several blood cell types that are involved in the development of atherosclerosis, the role of platelets during the thrombotic occlusion of ruptured or eroded plaques is well established and clinically exploited as evident by the extensive use of platelet inhibitors. However, there is increasing evidence that platelets are also involved in the earlier stages of atheroma development by exhibiting pro-inflammatory activities. The scope of this review is to describe the role of platelets in the initiation and propagation stages of atherosclerosis and beyond; in atherothrombotic complications.
Collapse
Affiliation(s)
- Daniëlle M Coenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Alexandra C A Heinzmann
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Mieke F A Karel
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Judith M E M Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Rory R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
62
|
Nocella C, Mezzaroma I, Cammisotto V, Castellani V, Milito C, Rugova A, Frati G, Pignatelli P, Violi F, Pastori D, Carnevale R. Lipopolysaccharide induces platelet activation in HIV patients: the role of different viral load patterns. HIV Med 2021; 22:434-444. [PMID: 33426758 DOI: 10.1111/hiv.13059] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/26/2020] [Accepted: 12/12/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVES This study aimed to assess whether gut-derived lipopolysaccharide (LPS) could affect platelet function in HIV-1 patients with residual viral load. METHODS In 23 HIV-1 patients on effective antiretroviral treatment, 10 treatment-naïve HIV-1 subjects and 20 healthy subjects (HS), LPS, zonulin, markers of platelet activation and oxidative stress were evaluated. In vitro, platelets from HS were exposed to plasma from HIV-1-infected treated and untreated patients. RESULTS Compared with HS, LPS was higher in treated and treatment-naïve subjects with HIV-1 (7.7 ± 2.9, 80.9 ± 13.7 and 75.3 ± 22.6 pg/mL, P < 0.001 vs. HS) as well as serum zonulin (1.3 ± 0.5, 6.1 ± 1.5 and 5.3 ± 1.7 ng/mL, P < 0.001 vs. HS). LPS and zonulin were correlated in HIV patients (Spearman correlation coefficient (rS) = 0.73, P < 0.0001). Levels of soluble CD40 ligand (sCD40L), soluble P-selectin (sP-selectin) and thromboxane B2 (TxB2 ) were higher in HIV-1-treated and treatment-naïve subjects compared with HS as well as NADPH oxidase 2 (NOX2) activation and hydrogen peroxide (H2 O2 ) production. In vitro, sCD40L, sP-selectin and TxB2 production, NOX2 activation and p47phox phosphorylation were higher in platelets exposed to plasma from HIV-1 patients with different viral load compared with the exposure to plasma from HS. This effect was blunted in platelets pre-treated with TLR4 or TLR7 inhibitors. CONCLUSIONS Low-grade endotoxaemia and persistent viraemia increase platelet function with a mechanism mediated by NOX2 in patients with HIV-1 infection.
Collapse
Affiliation(s)
- Cristina Nocella
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Ivano Mezzaroma
- Department of Clinical Medicine, Sapienza University of Rome, Rome, Italy
| | - Vittoria Cammisotto
- Department of General Surgery and Surgical Specialty Paride Stefanini, Sapienza University of Rome, Rome, Italy
| | - Valentina Castellani
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Cinzia Milito
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alban Rugova
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy
| | - Pasquale Pignatelli
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy.,Mediterranea, Cardiocentro, Napoli, Italy
| | - Francesco Violi
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy.,Mediterranea, Cardiocentro, Napoli, Italy
| | - Daniele Pastori
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Roberto Carnevale
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,Mediterranea, Cardiocentro, Napoli, Italy
| |
Collapse
|
63
|
Vallance TM, Sheard JJ, Meng Y, Torre EC, Patel K, Widera D, Vaiyapuri S. Development and characterization of a novel, megakaryocyte NF-κB reporter cell line for investigating inflammatory responses. J Thromb Haemost 2021; 19:107-120. [PMID: 33037735 DOI: 10.1111/jth.15118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/27/2020] [Accepted: 09/24/2020] [Indexed: 11/27/2022]
Abstract
Essentials An easily detectable readout in megakaryocyte cell lines will enhance inflammatory research in these cells. Here, we report the development and characterization of a novel megakaryocyte NF-κB-reporter cell line (Meg-01R). Multiple inflammatory molecules modulate NF-κB activity in Meg-01R cells. Meg-01R cells respond to small molecule inhibitors such as IMD0354 and C87 that are known to inhibit NF-κB activity upon stimulation with TNFα. ABSTRACT: Background Because of the difficulties in acquiring large numbers of megakaryocytes, the impact of inflammatory responses on these cells and their ability to produce fully functional platelets under various pathological conditions has not been investigated in detail. Objectives The primary objective of this study is to develop and functionally characterize a novel megakaryocyte nuclear factor κB (NF-κB) reporter cell line to determine the effects of various inflammatory molecules on megakaryocytes and their signalling pathways. Methods A Meg-01-NF-κB-GFP-Luc (Meg-01R) cell line was developed by inserting a reporter NF-κB-GFP-Luc cassette into normal Meg-01 cells to produce luciferase following activation of NF-κB to enable easy detection of pro-inflammatory and reparative signalling. Results and conclusions Meg-01 and Meg-01R cells have comparable characteristics, including the expression of both GPIbα and integrin β3 . Meg-01R cells responded to various inflammatory molecules as measured by NF-κB-dependent bioluminescence. For example, inflammatory molecules such as tumor necrosis factor-α and Pam3CSK4 increased NF-κB activity, whereas an antimicrobial peptide, LL37, reduced its activity. Meg-01R cells were also found to be sensitive to inhibitors (IMD0354 and C87) of inflammatory pathways. Notably, Meg-01R cells were able to respond to lipopolysaccharide (LPS; non-ultrapure), although it was not able to react to ultrapure LPS because of the lack of sufficient TLR4 molecules on their surface. For the first time, we report the development and characterization of a novel megakaryocyte NF-κB reporter cell line (Meg-01R) as a robust tool to study the inflammatory responses/signalling of megakaryocytes upon stimulation with a broad range of inflammatory molecules that can affect NF-κB activity.
Collapse
Affiliation(s)
| | | | - Yiming Meng
- School of Pharmacy, University of Reading, Reading, UK
| | | | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, UK
| | - Darius Widera
- School of Pharmacy, University of Reading, Reading, UK
| | | |
Collapse
|
64
|
Shannon O. The role of platelets in sepsis. Res Pract Thromb Haemost 2021; 5:27-37. [PMID: 33537527 PMCID: PMC7845078 DOI: 10.1002/rth2.12465] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/06/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022] Open
Abstract
A State of the Art lecture titled "The role of platelets in sepsis" was presented at the ISTH congress in 2020. Sepsis is a life-threatening organ dysfunction caused by a dysregulated and multifaceted host response to infection. Platelets play a significant role in the coordinated immune response to infection and therefore in the inflammation and coagulation dysfunction that contributes to organ damage in sepsis. Thrombocytopenia has a high incidence in sepsis, and it is a marker of poor prognosis. The genesis of thrombocytopenia is likely multifactorial, and unraveling the involved molecular mechanisms will allow development of biomarkers of platelet function in sepsis. Such platelet biomarkers can facilitate study of antiplatelet interventions as immunomodulatory treatment in sepsis. Finally, relevant new data on this topic presented during the 2020 ISTH virtual congress are reviewed.
Collapse
Affiliation(s)
- Oonagh Shannon
- Division of Infection MedicineDepartment of Clinical SciencesFaculty of MedicineLund UniversityLundSweden
| |
Collapse
|
65
|
Bang J, Jeon WK. Mumefural Improves Blood Flow in a Rat Model of FeCl 3-Induced Arterial Thrombosis. Nutrients 2020; 12:nu12123795. [PMID: 33322041 PMCID: PMC7763683 DOI: 10.3390/nu12123795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 01/04/2023] Open
Abstract
Mumefural (MF), a bioactive component of the processed fruit of Prunus mume Sieb. et Zucc, is known to inhibit platelet aggregation induced by agonists in vitro. In this study, we investigated the anti-thrombotic effects of MF using a rat model of FeCl3-induced arterial thrombosis. Sprague–Dawley rats were intraperitoneally injected with MF (0.1, 1, or 10 mg/kg) 30 min before 35% FeCl3 treatment to measure the time to occlusion using a laser Doppler flowmeter and to assess the weight of the blood vessels containing thrombus. MF treatment significantly improved blood flow by inhibiting occlusion and thrombus formation. MF also prevented collagen fiber damage in injured vessels and inhibited the expression of the platelet activation-related proteins P-selectin and E-selectin. Moreover, MF significantly reduced the increased inflammatory signal of nuclear factor (NF)-κB, toll-like receptor 4 (TLR4), tumor necrosis factor (TNF)-α, and interleukin (IL)-6 in blood vessels. After administration, MF was detected in the plasma samples of rats with a bioavailability of 36.95%. Therefore, we suggest that MF may improve blood flow as a candidate component in dietary supplements for improving blood flow and preventing blood circulation disorders.
Collapse
Affiliation(s)
- Jihye Bang
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Korea;
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Korea
| | - Won Kyung Jeon
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Korea;
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Korea
- Correspondence: ; Tel.: +82-2-958-6992
| |
Collapse
|
66
|
Ghimire S, Ravi S, Budhathoki R, Arjyal L, Hamal S, Bista A, Khadka S, Uprety D. Current understanding and future implications of sepsis-induced thrombocytopenia. Eur J Haematol 2020; 106:301-305. [PMID: 33191517 DOI: 10.1111/ejh.13549] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023]
Abstract
Sepsis is a global health burden that needs intensive medical care. Thrombocytopenia in sepsis is well known to increase morbidity as well as mortality. Several studies have been performed both in animal models and in humans to understand the mechanism by which sepsis causes thrombocytopenia. Recent studies have shown that inhibiting thrombocytopenia improves outcomes in sepsis patients. Understanding these mechanisms to identify targets in use of newer treatment modalities besides using resuscitation measures, antibiotics and removal of thrombocytopenia inducing agent could potentially help us improve outcomes in sepsis.
Collapse
Affiliation(s)
- Subash Ghimire
- Department of Medicine, Guthrie Robert Packer Hospital, Sayre, PA, USA
| | - Swapna Ravi
- Department of Medicine, Guthrie Robert Packer Hospital, Sayre, PA, USA
| | - Rasmita Budhathoki
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Lubina Arjyal
- Department of Medicine, Gundersen Lutheran Medical Center, La Crosse, WI, USA
| | - Savyata Hamal
- Zainul Haque Sikder Women's Medical College, Bangladesh
| | - Amir Bista
- Department of Hematology-Oncology, Gundersen Lutheran Medical Center, LA Crosse, WI, USA
| | - Sushmita Khadka
- Department of Medicine, Guthrie Robert Packer Hospital, Sayre, PA, USA
| | - Dipesh Uprety
- Department of Hematology-Oncology, Gundersen Lutheran Medical Center, LA Crosse, WI, USA
| |
Collapse
|
67
|
Scheuermann L, Pei G, Domaszewska T, Zyla J, Oberbeck-Müller D, Bandermann S, Feng Y, Ruiz Moreno JS, Opitz B, Mollenkopf HJ, Kaufmann SHE, Dorhoi A. Platelets Restrict the Oxidative Burst in Phagocytes and Facilitate Primary Progressive Tuberculosis. Am J Respir Crit Care Med 2020; 202:730-744. [PMID: 32421376 DOI: 10.1164/rccm.201910-2063oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Rationale: Platelets are generated in the capillaries of the lung, control hemostasis, and display immunological functions. Tuberculosis primarily affects the lung, and patients show platelet changes and hemoptysis. A role of platelets in immunopathology of pulmonary tuberculosis requires careful assessment.Objectives: To identify the dynamics and interaction partners of platelets in the respiratory tissue and establish their impact on the outcome of pulmonary tuberculosis.Methods: Investigations were primarily performed in murine models of primary progressive pulmonary tuberculosis, by analysis of mouse strains with variable susceptibility to Mycobacterium tuberculosis infection using platelet depletion and delivery of antiplatelet drugs.Measurements and Main Results: Platelets were present at the site of infection and formed aggregates with different myeloid subsets during experimental tuberculosis. Such aggregates were also detected in patients with tuberculosis. Platelets were detrimental during the early phase of infection, and this effect was uncoupled from their canonical activation. Platelets left lung cell dynamics and patterns of antimycobacterial T-cell responses unchanged but hampered antimicrobial defense by restricting production of reactive oxygen species in lung-residing myeloid cells.Conclusions: Platelets are detrimental in primary progressive pulmonary tuberculosis, orchestrate lung immunity by modulating innate immune responsiveness, and may be amenable to new interventions for this deadly disease.
Collapse
Affiliation(s)
| | - Gang Pei
- Immunology Department and.,Institute of Immunology, Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany
| | | | - Joanna Zyla
- Immunology Department and.,Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| | | | | | - Yonghong Feng
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Juan Sebastian Ruiz Moreno
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Bastian Opitz
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Stefan H E Kaufmann
- Immunology Department and.,Hagler Institute for Advanced Study at Texas A&M University, College Station, Texas; and
| | - Anca Dorhoi
- Immunology Department and.,Institute of Immunology, Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany.,Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany
| |
Collapse
|
68
|
The involvement of toll-like receptors 2 and 4 in human platelet signalling pathways. Cell Signal 2020; 76:109817. [PMID: 33132157 DOI: 10.1016/j.cellsig.2020.109817] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 01/01/2023]
Abstract
In addition to haemostasis, platelets play an essential role in mechanisms of inflammation and in immunological reactions. Platelets express various toll-like receptors (TLR) on their surface, among them TLR2 and TLR4, which are important for the recognition of bacterial patterns. This study compared TLR2- and TLR4-dependent platelet signalling and their effect on platelet function. Platelet-rich-plasma and washed platelets were prepared from peripheral blood samples of healthy donors. Pam3CSK4 or LPS (lipopolysaccharides from Escherichia coli) were used for stimulation of TLR2 and TLR4. Intracellular signalling pathways were investigated by Western blot. TLR2- and TLR4-mediated specific transcription factor DNA binding activity was measured by the nuclear factor kappa B (NFκB) transcription factor assay kit. Platelet adhesion and glycoprotein Ib function were assessed by immunofluorescence staining and analysis of ristocetin-induced agglutination. Both, Pam3CSK4 and LPS were able to induce NFκB-mediated and classical activating platelet signalling with a higher stimulatory capacity of TLR2. In addition, TLR2 and TLR4 activation led to a similar activation of inhibitory pathways. In contrast to TLR2, stimulation of TLR4 resulted in decreased Akt/protein kinase B phosphorylation conditioned by enhanced protein phosphatase 2A activity. TLR4-mediated signalling induced platelet adhesion and facilitated ristocetin-induced platelet agglutination. In conclusion, Pam3CSK4 directly induces aggregation via classical activation cascades, whereas LPS enhances platelet adhesion and glycoprotein receptor Ib-dependent platelet agglutination.
Collapse
|
69
|
Portier I, Campbell RA. Role of Platelets in Detection and Regulation of Infection. Arterioscler Thromb Vasc Biol 2020; 41:70-78. [PMID: 33115274 DOI: 10.1161/atvbaha.120.314645] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Platelets are classically known as essential mediators of hemostasis and thrombosis. However, in recent years, platelets have gained recognition for their inflammatory functions, which modulate the immune response during infectious diseases. Platelets contain various immunoreceptors that enable them to act as sentinels to recognize intravascular pathogens. Upon activation, platelets directly limit pathogen growth through the release of AMPs (antimicrobial proteins) and ensure pathogen clearance through activation of immune cells. However, aberrant platelet activation can lead to inflammation and thrombotic events.
Collapse
Affiliation(s)
- Irina Portier
- University of Utah Molecular Medicine Program, Salt Lake City (I.P., R.A.C.)
| | - Robert A Campbell
- University of Utah Molecular Medicine Program, Salt Lake City (I.P., R.A.C.).,Department of Internal Medicine, University of Utah, Salt Lake City (R.A.C.)
| |
Collapse
|
70
|
Gautam I, Storad Z, Filipiak L, Huss C, Meikle CK, Worth RG, Wuescher LM. From Classical to Unconventional: The Immune Receptors Facilitating Platelet Responses to Infection and Inflammation. BIOLOGY 2020; 9:E343. [PMID: 33092021 PMCID: PMC7589078 DOI: 10.3390/biology9100343] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/06/2020] [Accepted: 10/15/2020] [Indexed: 12/14/2022]
Abstract
Platelets have long been recognized for their role in maintaining the balance between hemostasis and thrombosis. While their contributions to blood clotting have been well established, it has been increasingly evident that their roles extend to both innate and adaptive immune functions during infection and inflammation. In this comprehensive review, we describe the various ways in which platelets interact with different microbes and elicit immune responses either directly, or through modulation of leukocyte behaviors.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Leah M. Wuescher
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (I.G.); (Z.S.); (L.F.); (C.H.); (C.K.M.); (R.G.W.)
| |
Collapse
|
71
|
Heinzmann AC, Karel MF, Coenen DM, Vajen T, Meulendijks NM, Nagy M, Suylen DP, Cosemans JM, Heemskerk JW, Hackeng TM, Koenen RR. Complementary roles of platelet αIIbβ3 integrin, phosphatidylserine exposure and cytoskeletal rearrangement in the release of extracellular vesicles. Atherosclerosis 2020; 310:17-25. [DOI: 10.1016/j.atherosclerosis.2020.07.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/26/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022]
|
72
|
Maouia A, Rebetz J, Kapur R, Semple JW. The Immune Nature of Platelets Revisited. Transfus Med Rev 2020; 34:209-220. [PMID: 33051111 PMCID: PMC7501063 DOI: 10.1016/j.tmrv.2020.09.005] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 01/08/2023]
Abstract
Platelets are the primary cellular mediators of hemostasis and this function firmly acquaints them with a variety of inflammatory processes. For example, platelets can act as circulating sentinels by expressing Toll-like receptors (TLR) that bind pathogens and this allows platelets to effectively kill them or present them to cells of the immune system. Furthermore, activated platelets secrete and express many pro- and anti-inflammatory molecules that attract and capture circulating leukocytes and direct them to inflamed tissues. In addition, platelets can directly influence adaptive immune responses via secretion of, for example, CD40 and CD40L molecules. Platelets are also the source of most of the microvesicles in the circulation and these miniscule elements further enhance the platelet’s ability to communicate with the immune system. More recently, it has been demonstrated that platelets and their parent cells, the megakaryocytes (MK), can also uptake, process and present both foreign and self-antigens to CD8+ T-cells conferring on them the ability to directly alter adaptive immune responses. This review will highlight several of the non-hemostatic attributes of platelets that clearly and rightfully place them as integral players in immune reactions. Platelets can act as circulating sentinels by expressing pathogen-associated molecular pattern receptors that bind pathogens and induce their killing and elimination. Activated platelets secrete and express a multitude of pro- and anti-inflammatory molecules that attract and capture circulating leukocytes and direct them to inflamed tissues. Platelets express and secrete many critical immunoregulatory molecules that significantly affect both innate and adaptive immune responses. Platelets are the primary source of microparticles in the circulation and these augment the platelet’s ability to communicate with the immune system. Platelets and megakaryocytes can act as antigen presenting cells and present both foreign- and self-peptides to T-cells.
Collapse
Affiliation(s)
- Amal Maouia
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Johan Rebetz
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Rick Kapur
- Sanquin Research, Department of Experimental Immunohematology, Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - John W Semple
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden; Clinical Immunology and Transfusion Medicine, Office of Medical Services, Region Skåne, Lund, Sweden.
| |
Collapse
|
73
|
Belizaire R, Makar RS. Non-Alloimmune Mechanisms of Thrombocytopenia and Refractoriness to Platelet Transfusion. Transfus Med Rev 2020; 34:242-249. [PMID: 33129606 PMCID: PMC7494440 DOI: 10.1016/j.tmrv.2020.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/24/2020] [Accepted: 09/03/2020] [Indexed: 12/11/2022]
Abstract
Refractoriness to platelet transfusion is a common clinical problem encountered by the transfusion medicine specialist. It is well recognized that most causes of refractoriness to platelet transfusion are not a consequence of alloimmunization to human leukocyte, platelet-specific, or ABO antigens, but are a consequence of platelet sequestration and consumption. This review summarizes the clinical factors that result in platelet refractoriness and highlights recent data describing novel biological mechanisms that contribute to this clinical problem.
Collapse
Affiliation(s)
- Roger Belizaire
- Associate Director, Adult Transfusion Medicine, Brigham and Women's Hospital, Boston, MA
| | - Robert S Makar
- Director, Blood Transfusion Service, Massachusetts General Hospital, Boston, MA.
| |
Collapse
|
74
|
Revisiting Platelets and Toll-Like Receptors (TLRs): At the Interface of Vascular Immunity and Thrombosis. Int J Mol Sci 2020; 21:ijms21176150. [PMID: 32858930 PMCID: PMC7504402 DOI: 10.3390/ijms21176150] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 12/19/2022] Open
Abstract
While platelet function has traditionally been described in the context of maintaining vascular integrity, recent evidence suggests that platelets can modulate inflammation in a much more sophisticated and nuanced manner than previously thought. Some aspects of this expanded repertoire of platelet function are mediated via expression of Toll-like receptors (TLRs). TLRs are a family of pattern recognition receptors that recognize pathogen-associated and damage-associated molecular patterns. Activation of these receptors is crucial for orchestrating and sustaining the inflammatory response to both types of danger signals. The TLR family consists of 10 known receptors, and there is at least some evidence that each of these are expressed on or within human platelets. This review presents the literature on TLR-mediated platelet activation for each of these receptors, and the existing understanding of platelet-TLR immune modulation. This review also highlights unresolved methodological issues that potentially contribute to some of the discrepancies within the literature, and we also suggest several recommendations to overcome these issues. Current understanding of TLR-mediated platelet responses in influenza, sepsis, transfusion-related injury and cardiovascular disease are discussed, and key outstanding research questions are highlighted. In summary, we provide a resource—a “researcher’s toolkit”—for undertaking further research in the field of platelet-TLR biology.
Collapse
|
75
|
Dib PRB, Quirino-Teixeira AC, Merij LB, Pinheiro MBM, Rozini SV, Andrade FB, Hottz ED. Innate immune receptors in platelets and platelet-leukocyte interactions. J Leukoc Biol 2020; 108:1157-1182. [PMID: 32779243 DOI: 10.1002/jlb.4mr0620-701r] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/11/2020] [Accepted: 06/28/2020] [Indexed: 12/14/2022] Open
Abstract
Platelets are chief cells in hemostasis. Apart from their hemostatic roles, platelets are major inflammatory effector cells that can influence both innate and adaptive immune responses. Activated platelets have thromboinflammatory functions linking hemostatic and immune responses in several physiological and pathological conditions. Among many ways in which platelets exert these functions, platelet expression of pattern recognition receptors (PRRs), including TLR, Nod-like receptor, and C-type lectin receptor families, plays major roles in sensing and responding to pathogen-associated or damage-associated molecular patterns (PAMPs and DAMPs, respectively). In this review, an increasing body of evidence is compiled showing the participation of platelet innate immune receptors, including PRRs, in infectious diseases, sterile inflammation, and cancer. How platelet recognition of endogenous DAMPs participates in sterile inflammatory diseases and thrombosis is discussed. In addition, platelet recognition of both PAMPs and DAMPs initiates platelet-mediated inflammation and vascular thrombosis in infectious diseases, including viral, bacterial, and parasite infections. The study also focuses on the involvement of innate immune receptors in platelet activation during cancer, and their contribution to tumor microenvironment development and metastasis. Finally, how innate immune receptors participate in platelet communication with leukocytes, modulating leukocyte-mediated inflammation and immune functions, is highlighted. These cell communication processes, including platelet-induced release of neutrophil extracellular traps, platelet Ag presentation to T-cells and platelet modulation of monocyte cytokine secretion are discussed in the context of infectious and sterile diseases of major concern in human health, including cardiovascular diseases, dengue, HIV infection, sepsis, and cancer.
Collapse
Affiliation(s)
- Paula Ribeiro Braga Dib
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil.,Laboratory of Immunology, Infectious Diseases and Obesity, Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Anna Cecíllia Quirino-Teixeira
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Laura Botelho Merij
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Mariana Brandi Mendonça Pinheiro
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Stephane Vicente Rozini
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Fernanda Brandi Andrade
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Eugenio Damaceno Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| |
Collapse
|
76
|
Abstract
Platelets, small anucleate cells circulating in the blood, are critical mediators in haemostasis and thrombosis. Interestingly, recent studies demonstrated that platelets contain both pro-inflammatory and anti-inflammatory molecules, equipping platelets with immunoregulatory function in both innate and adaptive immunity. In the context of infectious diseases, platelets are involved in early detection of invading microorganisms and are actively recruited to sites of infection. Platelets exert their effects on microbial pathogens either by direct binding to eliminate or restrict dissemination, or by shaping the subsequent host immune response. Reciprocally, many invading microbial pathogens can directly or indirectly target host platelets, altering platelet count or/and function. In addition, microbial pathogens can impact the host auto- and alloimmune responses to platelet antigens in several immune-mediated diseases, such as immune thrombocytopenia, and fetal and neonatal alloimmune thrombocytopenia. In this review, we discuss the mechanisms that contribute to the bidirectional interactions between platelets and various microbial pathogens, and how these interactions hold relevant implications in the pathogenesis of many infectious diseases. The knowledge obtained from "well-studied" microbes may also help us understand the pathogenesis of emerging microbes, such as SARS-CoV-2 coronavirus.
Collapse
Affiliation(s)
- Conglei Li
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto Platelet Immunobiology Group, University of Toronto, Toronto, ON, Canada
| | - June Li
- Toronto Platelet Immunobiology Group, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON, Canada
| | - Heyu Ni
- Toronto Platelet Immunobiology Group, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
77
|
Chen Y, Zhong H, Zhao Y, Luo X, Gao W. Role of platelet biomarkers in inflammatory response. Biomark Res 2020; 8:28. [PMID: 32774856 PMCID: PMC7397646 DOI: 10.1186/s40364-020-00207-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 07/23/2020] [Indexed: 12/20/2022] Open
Abstract
Beyond hemostasis, thrombosis and wound healing, it is becoming increasingly clear that platelets play an integral role in inflammatory response and immune regulation. Platelets recognize pathogenic microorganisms and secrete various immunoregulatory cytokines and chemokines, thus facilitating a variety of immune effects and regulatory functions. In this review, we discuss recent advances in signaling of platelet activation-related biomarkers in inflammatory settings and application prospects to apply for disease diagnosis and treatment.
Collapse
Affiliation(s)
- Yufei Chen
- Department of Cardiology, Huashan Hospital, Fudan University, No.12 Middle Wulumuqi Road, Jing'an District, Shanghai, 200040 China
| | - Haoxuan Zhong
- Department of Cardiology, Huashan Hospital, Fudan University, No.12 Middle Wulumuqi Road, Jing'an District, Shanghai, 200040 China
| | - Yikai Zhao
- Department of Cardiology, Huashan Hospital, Fudan University, No.12 Middle Wulumuqi Road, Jing'an District, Shanghai, 200040 China
| | - Xinping Luo
- Department of Cardiology, Huashan Hospital, Fudan University, No.12 Middle Wulumuqi Road, Jing'an District, Shanghai, 200040 China
| | - Wen Gao
- Department of Cardiology, Huashan Hospital, Fudan University, No.12 Middle Wulumuqi Road, Jing'an District, Shanghai, 200040 China
| |
Collapse
|
78
|
How Fungal Glycans Modulate Platelet Activation via Toll-Like Receptors Contributing to the Escape of Candida albicans from the Immune Response. Antibiotics (Basel) 2020; 9:antibiotics9070385. [PMID: 32645848 PMCID: PMC7399910 DOI: 10.3390/antibiotics9070385] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/23/2020] [Accepted: 07/06/2020] [Indexed: 12/20/2022] Open
Abstract
Platelets are essential for vascular repair and for the maintenance of blood homeostasis. They contribute to the immune defence of the host against many infections caused by bacteria, viruses and fungi. Following infection, platelet function is modified, and these cells form aggregates with microorganisms leading, to a decrease in the level of circulating platelets. During candidaemia, mannans, β-glucans and chitin, exposed on the cell wall of Candida albicans, an opportunistic pathogenic yeast of humans, play an important role in modulation of the host response. These fungal polysaccharides are released into the circulation during infection and their detection allows the early diagnosis of invasive fungal infections. However, their role in the modulation of the immune response and, in particular, that of platelets, is not well understood. The structure and solubility of glycans play an important role in the orientation of the immune response of the host. This short review focuses on the effect of fungal β-glucans and chitin on platelet activation and how these glycans modulate platelet activity via Toll-like receptors, contributing to the escape of C. albicans from the immune response.
Collapse
|
79
|
Banerjee M, Huang Y, Joshi S, Popa GJ, Mendenhall MD, Wang QJ, Garvy BA, Myint T, Whiteheart SW. Platelets Endocytose Viral Particles and Are Activated via TLR (Toll-Like Receptor) Signaling. Arterioscler Thromb Vasc Biol 2020; 40:1635-1650. [PMID: 32434410 PMCID: PMC7316618 DOI: 10.1161/atvbaha.120.314180] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Thrombocytopenia is associated with many viral infections suggesting virions interact with and affect platelets. Consistently, viral particles are seen inside platelets, and platelet activation markers are detected in viremic patients. In this article, we sought mechanistic insights into these virion/platelet interactions by examining how platelets endocytose, traffic, and are activated by a model virion. Approach and Results: Using fluorescently tagged HIV-1 pseudovirions, 3-dimensional structured illumination microscopy, and transgenic mouse models, we probed the interactions between platelets and virions. Mouse platelets used known endocytic machinery, that is, dynamin, VAMP (vesicle-associated membrane protein)-3, and Arf6 (ADP-ribosylation factor 6), to take up and traffic HIV-1 pseudovirions. Endocytosed HIV-1 pseudovirions trafficked through early (Rab4+) and late endosomes (Rab7+), and then to an LC3+ (microtubule-associated protein 1A/1B-light chain 3) compartment. Incubation with virions induced IRAK4 (interleukin 1 receptor-associated kinase 4), Akt (protein kinase B), and IKK (IκB kinase) activation, granule secretion, and platelet-leukocyte aggregate formation. This activation required TLRs (Toll-like receptors) and MyD88 (myeloid differentiation primary response protein 88) but was less extensive and slower than activation with thrombin. In vivo, HIV-1 pseudovirions injection led to virion uptake and platelet activation, as measured by IKK activation, platelet-leukocyte aggregate formation, and mild thrombocytopenia. All were decreased in VAMP-3-/- and, megakaryocyte/platelet-specific, Arf6-/- mice. Similar platelet activation profiles (increased platelet-leukocyte aggregates, plasma platelet factor 4, and phospho-IκBα) were detected in newly diagnosed and antiretroviral therapy-controlled HIV-1+ patients. CONCLUSIONS Collectively, our data provide mechanistic insights into the cell biology of how platelets endocytose and process virions. We propose a mechanism by which platelets sample the circulation and respond to potential pathogens that they take up.
Collapse
Affiliation(s)
- Meenakshi Banerjee
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
| | - Yunjie Huang
- Division of Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Smita Joshi
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
- Lexington VA Health Care System, Lexington, KY
| | - Gabriel J. Popa
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
| | - Michael D. Mendenhall
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
| | - Qing Jun Wang
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY
| | - Beth A. Garvy
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY
| | - Thein Myint
- Department of Infectious Diseases, Bluegrass Care Clinic, Kentucky Clinic, University of Kentucky, Lexington, KY
| | - Sidney W. Whiteheart
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
- Lexington VA Health Care System, Lexington, KY
| |
Collapse
|
80
|
Zhou Y, Little PJ, Downey L, Afroz R, Wu Y, Ta HT, Xu S, Kamato D. The Role of Toll-like Receptors in Atherothrombotic Cardiovascular Disease. ACS Pharmacol Transl Sci 2020; 3:457-471. [PMID: 32566912 PMCID: PMC7296543 DOI: 10.1021/acsptsci.9b00100] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Indexed: 02/06/2023]
Abstract
Toll-like receptors (TLRs) are dominant components of the innate immune system. Activated by both pathogen-associated molecular patterns and damage-associated molecular patterns, TLRs underpin the pathology of numerous inflammation related diseases that include not only immune diseases, but also cardiovascular disease (CVD), diabetes, obesity, and cancers. Growing evidence has demonstrated that TLRs are involved in multiple cardiovascular pathophysiologies, such as atherosclerosis and hypertension. Specifically, a trial called the Canakinumab Anti-inflammatory Thrombosis Outcomes Study showed the use of an antibody that neutralizes interleukin-1β, reduces the recurrence of cardiovascular events, demonstrating inflammation as a therapeutic target and also the research value of targeting the TLR system in CVD. In this review, we provide an update of the interplay between TLR signaling, inflammatory mediators, and atherothrombosis, with an aim to identify new therapeutic targets for atherothrombotic CVD.
Collapse
Affiliation(s)
- Ying Zhou
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
| | - Peter J. Little
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
- Department
of Pharmacy, Xinhua College of Sun Yat-Sen
University, Tianhe District, Guangzhou, Guangdong Province 510520, China
| | - Liam Downey
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
| | - Rizwana Afroz
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
| | - Yuao Wu
- Australian
Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, St Lucia, Queensland 4072, Australia
| | - Hang T. Ta
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
- Australian
Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, St Lucia, Queensland 4072, Australia
| | - Suowen Xu
- Aab
Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, United States
| | - Danielle Kamato
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
- Department
of Pharmacy, Xinhua College of Sun Yat-Sen
University, Tianhe District, Guangzhou, Guangdong Province 510520, China
| |
Collapse
|
81
|
The dual FXa/thrombin inhibitor SATI prevents fibrin and platelet deposition in hypercoagulant rats. Thromb Res 2020; 193:15-21. [PMID: 32505079 DOI: 10.1016/j.thromres.2020.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/23/2020] [Accepted: 05/12/2020] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Systemic hypercoagulation is often a severe complication of infective and inflammatory diseases, which overcome the hemostatic balance and lead to multiple thrombotic occlusions in the microvasculature and organ damage and is related to high mortality rates. SATI is a potent dual inhibitor of FXa and thrombin with antithrombotic efficacy in venous and arterial thrombosis models. In this study, the antithrombotic efficacy of SATI was investigated in a microthrombosis model in rats with an induced hypercoagulant state. MATERIALS AND METHODS The hypercoagulant state was generated by infusion of TF in sixty rats (12 groups, consisting of 5 rats each). SATI was administered in two different doses by constant infusion and its antithrombotic efficacy was investigated using two different approaches: 1) measuring 125I-fibrin deposition in various organs and 2) continuous whole-body imaging of 111In-platelet biodistribution in anesthetized animals. RESULTS After start of the TF infusion in rats with radioactively-labeled fibrinogen, the radioactivity was accumulated in liver, spleen, kidney, and mostly in the lung as a consequence of fibrin generation. SATI efficiently reduced the pulmonary deposition of fibrin in a dose- and time-dependent manner. In the SATI groups the splenic and renal radioactivity was enhanced at later time points probably as consequence of the clearance of 125I-fibrin(ogen). Imaging of rats that received 111In-platelets prior to systemic TF administration showed retention of the radioactivity mainly in the lungs in the control group. SATI efficiently blocked the platelet accumulation in the lungs and increased platelet recruitment by the spleen. CONCLUSIONS SATI is a promising candidate for prevention of microcirculatory disturbances by inhibiting fibrin deposition and platelet accumulation in the lungs and thereby conferring organ protection. Both methods used in this study are suitable for investigating the antithrombotic efficacy of new drugs in microthrombosis. Continuous imaging of 111In-platelets allowed for follow-up of thrombus formation in living animals without the need for tissue harvesting.
Collapse
|
82
|
Page MJ, Pretorius E. A Champion of Host Defense: A Generic Large-Scale Cause for Platelet Dysfunction and Depletion in Infection. Semin Thromb Hemost 2020; 46:302-319. [PMID: 32279287 PMCID: PMC7339151 DOI: 10.1055/s-0040-1708827] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Thrombocytopenia is commonly associated with sepsis and infections, which in turn are characterized by a profound immune reaction to the invading pathogen. Platelets are one of the cellular entities that exert considerable immune, antibacterial, and antiviral actions, and are therefore active participants in the host response. Platelets are sensitive to surrounding inflammatory stimuli and contribute to the immune response by multiple mechanisms, including endowing the endothelium with a proinflammatory phenotype, enhancing and amplifying leukocyte recruitment and inflammation, promoting the effector functions of immune cells, and ensuring an optimal adaptive immune response. During infection, pathogens and their products influence the platelet response and can even be toxic. However, platelets are able to sense and engage bacteria and viruses to assist in their removal and destruction. Platelets greatly contribute to host defense by multiple mechanisms, including forming immune complexes and aggregates, shedding their granular content, and internalizing pathogens and subsequently being marked for removal. These processes, and the nature of platelet function in general, cause the platelet to be irreversibly consumed in the execution of its duty. An exaggerated systemic inflammatory response to infection can drive platelet dysfunction, where platelets are inappropriately activated and face immunological destruction. While thrombocytopenia may arise by condition-specific mechanisms that cause an imbalance between platelet production and removal, this review evaluates a generic large-scale mechanism for platelet depletion as a repercussion of its involvement at the nexus of responses to infection.
Collapse
Affiliation(s)
- Martin J Page
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
83
|
Cleary SJ, Hobbs C, Amison RT, Arnold S, O'Shaughnessy BG, Lefrançais E, Mallavia B, Looney MR, Page CP, Pitchford SC. LPS-induced Lung Platelet Recruitment Occurs Independently from Neutrophils, PSGL-1, and P-Selectin. Am J Respir Cell Mol Biol 2020; 61:232-243. [PMID: 30768917 DOI: 10.1165/rcmb.2018-0182oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Platelets are recruited to inflammatory foci and contribute to host defense and inflammatory responses. Compared with platelet recruitment in hemostasis and thrombosis, the mechanisms of platelet recruitment in inflammation and host defense are poorly understood. Neutrophil recruitment to lung airspaces after inhalation of bacterial LPS requires platelets and PSGL-1 in mice. Given this association between platelets and neutrophils, we investigated whether recruitment of platelets to lungs of mice after LPS inhalation was dependent on PSGL-1, P-selectin, or interaction with neutrophils. BALB/c mice were administered intranasal LPS (O55:B5, 5 mg/kg) and, 48 hours later, lungs were collected and platelets and neutrophils quantified in tissue sections by immunohistochemistry. The effects of functional blocking antibody treatments targeting the platelet-neutrophil adhesion molecules, P-selectin or PSGL-1, or treatment with a neutrophil-depleting antibody targeting Ly6G, were tested on the extent of LPS-induced lung platelet recruitment. Separately in Pf4-Cre × mTmG mice, two-photon intravital microscopy was used to image platelet adhesion in live lungs. Inhalation of LPS caused both platelet and neutrophil recruitment to the lung vasculature. However, decreasing lung neutrophil recruitment by blocking PSGL-1, P-selectin, or depleting blood neutrophils had no effect on lung platelet recruitment. Lung intravital imaging revealed increased adhesion of platelets in the lung microvasculature which was not associated with thrombus formation. In conclusion, platelet recruitment to lungs in response to LPS occurs through mechanisms distinct from those mediating neutrophil recruitment, or the occurrence of pulmonary emboli.
Collapse
Affiliation(s)
- Simon J Cleary
- 1Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| | - Carl Hobbs
- 2the Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom; and
| | - Richard T Amison
- 1Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| | - Stephanie Arnold
- 1Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| | - Blaze G O'Shaughnessy
- 1Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| | - Emma Lefrançais
- 3Department of Medicine, University of California San Francisco, San Francisco, California
| | - Beñat Mallavia
- 3Department of Medicine, University of California San Francisco, San Francisco, California
| | - Mark R Looney
- 3Department of Medicine, University of California San Francisco, San Francisco, California
| | - Clive P Page
- 1Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| | - Simon C Pitchford
- 1Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| |
Collapse
|
84
|
Hasan RA, Koh AY, Zia A. The gut microbiome and thromboembolism. Thromb Res 2020; 189:77-87. [PMID: 32192995 DOI: 10.1016/j.thromres.2020.03.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/09/2020] [Accepted: 03/05/2020] [Indexed: 02/06/2023]
Abstract
The gut microbiome plays a critical role in various inflammatory conditions, and its modulation is a potential treatment option for these conditions. The role of the gut microbiome in the pathogenesis of thromboembolism has not been fully elucidated. In this review, we summarize the evidence linking the gut microbiome to the pathogenesis of arterial and venous thrombosis. In a human host, potentially pathogenic bacteria are normal residents of the human gut microbiome, but significantly outnumbered by commensal anaerobic bacteria. Several disease states with an increased risk of venous thromboembolism (VTE) are associated with an imbalance in the gut microbiome characterized by a decrease in commensal anaerobic bacteria and an increase in the abundance of pathogenic bacteria of which the most common is the gram-negative Enterobacteriaceae (ENTERO) family. Bacterial lipopolysaccharides (LPS), the glycolipids found on the outer membrane of gram-negative bacteria, is one of the links between the microbiome and hypercoagulability. LPS binds to toll-like receptors to activate endothelial cells and platelets, leading to activation of the coagulation cascade. Bacteria in the microbiome can also metabolite compounds in the diet to produce important metabolites like trimethylamine-N-oxide (TMAO). TMAO causes platelet hyperreactivity, promotes thrombus formation and is associated with cardiovascular disease. Modulating the gut microbiome to target LPS and TMAO levels may be an innovative approach for decreasing the risk of thrombosis.
Collapse
Affiliation(s)
- Rida Abid Hasan
- Department of Pediatrics, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Andrew Y Koh
- Department of Pediatrics, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, TX, United States of America; Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, United States of America; Harold C. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Ayesha Zia
- Department of Pediatrics, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, TX, United States of America.
| |
Collapse
|
85
|
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease in which a variety of circulating pro-inflammatory cells and dysregulated molecules are involved in disease aetiology and progression. Platelets are an important cellular element in the circulation that can bind several dysregulated molecules (such as collagen, thrombin and fibrinogen) that are present both in the synovium and the circulation of patients with RA. Platelets not only respond to dysregulated molecules in their environment but also transport and express their own inflammatory mediators, and serve as regulators at the boundary between haemostasis and immunity. Activated platelets also produce microparticles, which further convey signalling molecules and receptors to the synovium and circulation, thereby positioning these platelet-derived particles as strategic regulators of inflammation. These diverse functions come together to make platelets facilitators of cellular crosstalk in RA. Thus, the receptor functions, ligand binding potential and dysregulated signalling pathways in platelets are becoming increasingly important for treatment in RA. This Review aims to highlight the role of platelets in RA and the need to closely examine platelets as health indicators when designing effective pharmaceutical targets in this disease.
Collapse
|
86
|
Reduced miR-26b Expression in Megakaryocytes and Platelets Contributes to Elevated Level of Platelet Activation Status in Sepsis. Int J Mol Sci 2020; 21:ijms21030866. [PMID: 32013235 PMCID: PMC7036890 DOI: 10.3390/ijms21030866] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/22/2020] [Accepted: 01/26/2020] [Indexed: 12/16/2022] Open
Abstract
In sepsis, platelets may become activated via toll-like receptors (TLRs), causing microvascular thrombosis. Megakaryocytes (MKs) also express these receptors; thus, severe infection may modulate thrombopoiesis. To explore the relevance of altered miRNAs in platelet activation upon sepsis, we first investigated sepsis-induced miRNA expression in platelets of septic patients. The effect of abnormal Dicer level on miRNA expression was also evaluated. miRNAs were profiled in septic vs. normal platelets using TaqMan Open Array. We validated platelet miR-26b with its target SELP (P-selectin) mRNA levels and correlated them with clinical outcomes. The impact of sepsis on MK transcriptome was analyzed in MEG-01 cells after lipopolysaccharide (LPS) treatment by RNA-seq. Sepsis-reduced miR-26b was further studied using Dicer1 siRNA and calpain inhibition in MEG-01 cells. Out of 390 platelet miRNAs detected, there were 121 significantly decreased, and 61 upregulated in sepsis vs. controls. Septic platelets showed attenuated miR-26b, which were associated with disease severity and mortality. SELP mRNA level was elevated in sepsis, especially in platelets with increased mean platelet volume, causing higher P-selectin expression. Downregulation of Dicer1 generated lower miR-26b with higher SELP mRNA, while calpeptin restored miR-26b in MEG-01 cells. In conclusion, decreased miR-26b in MKs and platelets contributes to an increased level of platelet activation status in sepsis.
Collapse
|
87
|
Jerez-Dolz D, Torramade-Moix S, Palomo M, Moreno-Castaño A, Lopez-Vilchez I, Hernandez R, Badimon JJ, Zafar MU, Diaz-Ricart M, Escolar G. Internalization of microparticles by platelets is partially mediated by toll-like receptor 4 and enhances platelet thrombogenicity. Atherosclerosis 2019; 294:17-24. [PMID: 31945614 DOI: 10.1016/j.atherosclerosis.2019.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/22/2019] [Accepted: 12/19/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND AIMS Circulating platelet microparticles (PMP) are the most abundant in bloodstream, are highly procoagulant and contribute to cross-talk with inflammatory cells. The aim of the present study was to investigate the interactions of PMP with platelets and explore the involvement of toll-like receptor 4 (TLR-4). METHODS PMP were separated by ultracentrifugation of expired platelet concentrates and added to: i) washed platelets, to confirm uptake, by flow cytometry and confocal and transmission electron microscopy, ii) platelet rich plasma (PRP), to assess changes in platelet function due to uptake by aggregometry in response to ADP; and iii) whole blood, to evaluate heterotypic aggregate (HA) formation by flow cytometry. Moreover, whole blood previously enriched with platelets with internalized PMP was used to explore modifications in thromboelastometry parameters (ROTEM). The inhibitory action of anti-TLR-4 was investigated. RESULTS Confocal and ultrastructural microscopy studies revealed PMP internalization by platelets. Flow cytometry showed PMP-platelet association (p < 0.01 vs controls, at different PMP dilutions). PMP, at 1/20 dilution, increased HA (p < 0.05 vs controls), the percentage of maximal platelet aggregation to ADP (p < 0.05 vs controls), and accelerated clotting and clot formation times (p < 0.05 vs controls). Incubation of platelets with anti-TLR-4 prior to exposure to PMP reduced PMP-platelet association (p < 0.05 vs absence of the antibody), prevented HA formation, reduced maximal platelet aggregation and normalized ROTEM parameters. CONCLUSIONS Platelets exhibit internalization ability towards their own PMP, a process that potentiates their thrombogenicity and is partially mediated by the innate immunity receptor TLR-4.
Collapse
Affiliation(s)
- Didac Jerez-Dolz
- Hematopathology, Pathological Anatomy, Hospital Clinic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Sergi Torramade-Moix
- Hematopathology, Pathological Anatomy, Hospital Clinic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Marta Palomo
- Hematopathology, Pathological Anatomy, Hospital Clinic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain; Barcelona Endothelium Team, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain
| | - Ana Moreno-Castaño
- Hematopathology, Pathological Anatomy, Hospital Clinic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Irene Lopez-Vilchez
- Hematopathology, Pathological Anatomy, Hospital Clinic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Rosa Hernandez
- Hematopathology, Pathological Anatomy, Hospital Clinic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Juan Jose Badimon
- Atherothrombosis Research Unit, Icahn School of Medicine at Mount Sinai, New York, USA
| | - M Urooj Zafar
- Atherothrombosis Research Unit, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Maribel Diaz-Ricart
- Hematopathology, Pathological Anatomy, Hospital Clinic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; Barcelona Endothelium Team, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain
| | - Gines Escolar
- Hematopathology, Pathological Anatomy, Hospital Clinic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; Atherothrombosis Research Unit, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
88
|
Li Y, Ryan J, Xu F, Vostal JG. Macrophage Depletion Mitigates Platelet Aggregate Formation in Splenic Marginal Zone and Alleviates LPS-Associated Thrombocytopenia in Rats. Front Med (Lausanne) 2019; 6:300. [PMID: 31921873 PMCID: PMC6927931 DOI: 10.3389/fmed.2019.00300] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 11/29/2019] [Indexed: 11/19/2022] Open
Abstract
Sepsis is often accompanied with thrombocytopenia partly due to platelet sequestration in the lung and liver. The spleen can store up to one-third of circulating platelets and can also significantly affect platelet transfusion outcomes by accumulating platelets. However, in sepsis, it is not clear whether there are platelet changes in the spleen which could contribute to sepsis-associated thrombocytopenia and also influence platelet transfusion outcomes. By using confocal microscopy, we examined endogenous rat platelets and infused human platelets in the spleen of severe combined immune deficient Rag2 KO rats which were injected intraperitoneally with lipopolysaccharide (LPS). LPS-injected Rag2 KO rats developed sepsis as indicated by increased TNFa, IL-6, IL-1b, and IL-10 levels and thrombocytopenia. Large platelet aggregates were observed in the spleen with majority located in the marginal zone and closely associated with CD169+ macrophages. Depletion of macrophages by clodrosome resulted in reduction of LPS-induced cytokine generation and alleviated LPS-induced thrombocytopenia. Macrophage depletion also remarkedly diminished large platelet aggregate formation in the splenic marginal zone but had less effect on those in red pulp. Infusion of human platelets into LPS-injected rats failed to raise platelet counts in the peripheral blood. In LPS-injected rat spleen, human platelets interacted with aggregated rat platelets in the marginal zone. In contrast, human platelets infused into control rats were located outside of splenic marginal zone. This study provides morphological evidence of platelet aggregates in the splenic marginal zone in sepsis which can interact with infused platelets and thus can contribute to platelet infusion refractoriness in sepsis. It indicates that macrophages play an important role in LPS-associated thrombocytopenia. It also suggests that CD169+ macrophages support platelet aggregate formation in the splenic marginal zone.
Collapse
Affiliation(s)
- Ying Li
- Laboratory of Cellular Hematology, Division of Blood Components and Devices, Office of Blood Research and Review, Food and Drug Administration, Silver Spring, MD, United States
| | - Johannah Ryan
- Laboratory of Cellular Hematology, Division of Blood Components and Devices, Office of Blood Research and Review, Food and Drug Administration, Silver Spring, MD, United States
| | - Fei Xu
- Laboratory of Cellular Hematology, Division of Blood Components and Devices, Office of Blood Research and Review, Food and Drug Administration, Silver Spring, MD, United States
| | - Jaroslav G Vostal
- Laboratory of Cellular Hematology, Division of Blood Components and Devices, Office of Blood Research and Review, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
89
|
Cleary SJ, Rauzi F, Smyth E, Correia A, Hobbs C, Emerson M, Page CP, Pitchford SC. Radiolabelling and immunohistochemistry reveal platelet recruitment into lungs and platelet migration into airspaces following LPS inhalation in mice. J Pharmacol Toxicol Methods 2019; 102:106660. [PMID: 31838234 DOI: 10.1016/j.vascn.2019.106660] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/21/2019] [Accepted: 12/01/2019] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Platelets are under investigation for their role in host defence and inflammatory lung diseases and have been demonstrated to be recruited to the lung. However, the mechanisms and consequences of platelet recruitment into lungs are poorly understood. We have utilised a murine model to investigate the mechanisms of platelet involvement in lung inflammation induced by intranasal administration of LPS. OBJECTIVES Our aim was to characterise lung platelet recruitment following LPS inhalation in mice using immunohistochemistry, and non-invasive and invasive radiolabelled platelet tracking techniques. RESULTS Intranasal administration of LPS caused an increase in lung platelet staining in lung tissue and elicited the recruitment of radiolabelled platelets into the lung. Prior to these responses in the lung, we observed an earlier decrease in blood platelet counts, temporally associated with platelet recruitment to the liver and spleen. Non-invasive measurements of thoracic radioactivity reflected changes in blood counts rather than extravascular lung platelet recruitment. However, both in situ counting of radiolabelled platelets and immunostaining for platelet surface markers showed LPS-induced increases in extravascular platelets into lung airspaces suggesting that some of the platelets recruited to the lung enter air spaces. CONCLUSIONS Intranasal administration of LPS activates the innate immune response which includes a fall in peripheral blood platelet counts with subsequent platelet recruitment to the lung, spleen and liver, measured by immunohistochemistry and radiolabelling techniques.
Collapse
Affiliation(s)
- S J Cleary
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, UK
| | - F Rauzi
- National Heart & Lung Institute, Imperial College London, London, UK
| | - E Smyth
- National Heart & Lung Institute, Imperial College London, London, UK
| | - A Correia
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, UK
| | - C Hobbs
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - M Emerson
- National Heart & Lung Institute, Imperial College London, London, UK
| | - C P Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, UK
| | - S C Pitchford
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, UK.
| |
Collapse
|
90
|
Vallance TM, Ravishankar D, Albadawi DAI, Layfield H, Sheard J, Vaiyapuri R, Dash P, Patel K, Widera D, Vaiyapuri S. Effect of ultrapure lipopolysaccharides derived from diverse bacterial species on the modulation of platelet activation. Sci Rep 2019; 9:18258. [PMID: 31796818 PMCID: PMC6890654 DOI: 10.1038/s41598-019-54617-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 11/18/2019] [Indexed: 11/09/2022] Open
Abstract
Platelets are small circulating blood cells that play essential roles in the maintenance of haemostasis via blood clotting. However, they also play critical roles in the regulation of innate immune responses. Inflammatory receptors, specifically Toll-like receptor (TLR)-4, have been reported to modify platelet reactivity. A plethora of studies have reported controversial functions of TLR4 in the modulation of platelet function using various chemotypes and preparations of its ligand, lipopolysaccharide (LPS). The method of preparation of LPS may explain these discrepancies however this is not fully understood. Hence, to determine the impact of LPS on platelet activation, we used ultrapure preparations of LPS from Escherichia coli (LPSEC), Salmonella minnesota (LPSSM), and Rhodobacter sphaeroides (LPSRS) and examined their actions under diverse experimental conditions in human platelets. LPSEC did not affect platelet activation markers such as inside-out signalling to integrin αIIbβ3 or P-selectin exposure upon agonist-induced activation in platelet-rich plasma or whole blood whereas LPSSM and LPSRS inhibited platelet activation under specific conditions at supraphysiological concentrations. Overall, our data demonstrate that platelet activation is not largely influenced by any of the ultrapure LPS chemotypes used in this study on their own except under certain conditions.
Collapse
Affiliation(s)
- Thomas M Vallance
- School of Pharmacy, University of Reading, Reading, RG6 6UB, United Kingdom
| | | | - Dina A I Albadawi
- School of Pharmacy, University of Reading, Reading, RG6 6UB, United Kingdom
| | - Harry Layfield
- School of Pharmacy, University of Reading, Reading, RG6 6UB, United Kingdom
| | - Jonathan Sheard
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, University of Reading, Reading, RG6 6UB, United Kingdom.,Sheard BioTech Ltd, 20-22, Wenlock Road, London, N1 7GU, United Kingdom
| | | | - Philip Dash
- School of Biological Sciences, University of Reading, Reading, RG6 6UB, United Kingdom
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, RG6 6UB, United Kingdom
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, University of Reading, Reading, RG6 6UB, United Kingdom
| | - Sakthivel Vaiyapuri
- School of Pharmacy, University of Reading, Reading, RG6 6UB, United Kingdom.
| |
Collapse
|
91
|
D'Atri LP, Rodríguez CS, Miguel CP, Pozner RG, Ortiz Wilczyñski JM, Negrotto S, Carrera-Silva EA, Heller PG, Schattner M. Activation of toll-like receptors 2 and 4 on CD34 + cells increases human megakaryo/thrombopoiesis induced by thrombopoietin. J Thromb Haemost 2019; 17:2196-2210. [PMID: 31397069 DOI: 10.1111/jth.14605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/07/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Platelet Toll-like receptor (TLR)2/4 are key players in amplifying the host immune response; however, their role in human megakaryo/thrombopoiesis has not yet been defined. OBJECTIVES We evaluated whether Pam3CSK4 or lipopolysaccharide (LPS), TLR2/4 ligands respectively, modulate human megakaryocyte development and platelet production. METHODS CD34+ cells from human umbilical cord were stimulated with LPS or Pam3CSK4 with or without thrombopoietin (TPO). RESULTS CD34+ cells and megakaryocytes express TLR2 and TLR4 at both RNA and protein level; however, direct stimulation of CD34+ cells with LPS or Pam3CSK4 had no effect on cell growth. Interestingly, both TLR ligands markedly increased TPO-induced CD34+ cell proliferation, megakaryocyte number and maturity, proplatelet and platelet production when added at day 0. In contrast, this synergism was not observed when TLR agonists were added 7 days after TPO addition. Interleukin-6 (IL-6) release was observed upon CD34+ or megakaryocyte stimulation with LPS or Pam3CSK4 but not with TPO and this effect was potentiated in combination with TPO. The increased proliferation and IL-6 production induced by TPO + LPS or Pam3CSK4 were suppressed by TLR2/4 or IL-6 neutralizing antibodies, as well as by PI3K/AKT and nuclear factor-κB inhibitors. Additionally, increased proplatelet and platelet production were associated with enhanced nuclear translocation of nuclear factor-E2. Finally, the supernatants of CD34+ cells stimulated with TPO+LPS-induced CFU-M colonies. CONCLUSIONS Our data suggest that the activation of TLR2 and TLR4 in CD34+ cells and megakaryocytes in the presence of TPO may contribute to warrant platelet provision during infection episodes by an autocrine IL-6 loop triggered by PI3K/NF-κB axes.
Collapse
Affiliation(s)
- Lina Paola D'Atri
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Camila Sofía Rodríguez
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Carolina Paula Miguel
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Roberto Gabriel Pozner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Juan Manuel Ortiz Wilczyñski
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Soledad Negrotto
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Eugenio Antonio Carrera-Silva
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Paula Graciela Heller
- Institute of Medical Research Dr. Alfredo Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
- Department of Hematology Research, National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Institute of Medical Research (IDIM), Buenos Aires, Argentina
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| |
Collapse
|
92
|
Garbin LC, Olver CS. Platelet-Rich Products and Their Application to Osteoarthritis. J Equine Vet Sci 2019; 86:102820. [PMID: 32067662 DOI: 10.1016/j.jevs.2019.102820] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 08/04/2019] [Accepted: 10/22/2019] [Indexed: 12/15/2022]
Abstract
Autologous platelet-rich plasma (PRP) is a biological preparation made from the patient's own plasma that contains a platelet concentration above the whole blood baseline. Owing to the release of growth factors and other cytokines after degranulation, platelets have a central role in inflammation and in different stages of the healing process. For this reason, PRP-derived products have been used to enhance healing of musculoskeletal injuries and modulate progression of inflammatory processes, including osteoarthritis (OA). Osteoarthritis is one of the main causes of musculoskeletal disabilities in horses, and currently, there is no effective treatment for this disease. Treatments that focus on the modulation of inflammation and disease progression offer new hope for OA. Platelet-rich plasma provides a more practical and accessible option of therapy compared to other forms of biological treatment (i.e., stem cell therapies) and is believed to induce the production of functional matrix. However, several factors related to PRP production, including methods of preparation and application, and intraindividual variability, lead to an inconsistent product, precluding reliable conclusions about its efficacy for clinical use. The aim of this study was to review the benefits related to the clinical use of PRP in OA as well as factors that influence its use, the limitations of this treatment, and future directions of PRP research and therapy.
Collapse
Affiliation(s)
- Livia Camargo Garbin
- Department of Clinical Veterinary Sciences, School of Veterinary Medicine, Faculty of Medical Sciences, University of West Indies, St. Augustine, Trinidad and Tobago, West Indies.
| | - Christine S Olver
- Veterinary Diagnostic Laboratory, Clinical Pathology Section, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| |
Collapse
|
93
|
Birnie E, Claushuis TAM, Koh GCKW, Limmathurotsakul D, Day NPJ, Roelofs JJTH, Ware J, Hou B, de Vos AF, van der Poll T, van 't Veer C, Wiersinga WJ. Thrombocytopenia Impairs Host Defense Against Burkholderia pseudomallei (Melioidosis). J Infect Dis 2019; 219:648-659. [PMID: 30312422 PMCID: PMC6350952 DOI: 10.1093/infdis/jiy541] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/26/2018] [Indexed: 01/10/2023] Open
Abstract
Background Infection with the gram-negative bacillus Burkholderia pseudomallei (melioidosis) is an important cause of pneumosepsis in Southeast Asia and has a mortality of up to 40%. We aimed to assess the role of platelets in the host response against B. pseudomallei infection. Methods Association between platelet counts and mortality was determined in 1160 patients with culture-proven melioidosis. Mice treated with (low- or high-dose) platelet-depleting antibody were inoculated intranasally with B. pseudomallei and killed. Additional studies using functional glycoprotein Ibα-deficient mice were conducted. Results Thrombocytopenia was present in 31% of patients at admission and predicted mortality in melioidosis patients even after adjustment for confounders. In our murine-melioidosis model, platelet counts decreased, and mice treated with a platelet-depleting antibody showed enhanced mortality and higher bacterial loads compared to mice with normal platelet counts. Low platelet counts had a modest impact on early-pulmonary neutrophil influx. Reminiscent of their role in hemostasis, platelet depletion impaired vascular integrity, resulting in early lung bleeding. Glycoprotein Ibα-deficient mice had reduced platelet counts during B. pseudomallei infection together with an impaired local host defense in the lung. Conclusions Thrombocytopenia predicts mortality in melioidosis patients and, during experimental melioidosis, platelets play a protective role in both innate immunity and vascular integrity.
Collapse
Affiliation(s)
- Emma Birnie
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Center (UMC), University of Amsterdam, The Netherlands
| | - Theodora A M Claushuis
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Center (UMC), University of Amsterdam, The Netherlands
| | - Gavin C K W Koh
- Department of Medicine, University of Cambridge, United Kingdom
| | - Direk Limmathurotsakul
- Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand.,Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Center for Tropical Medicine and Global Health, University of Oxford, United Kingdom
| | - Nicholas P J Day
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.,Center for Tropical Medicine and Global Health, University of Oxford, United Kingdom
| | - Joris J T H Roelofs
- Department of Pathology, Amsterdam UMC, University of Amsterdam, The Netherlands
| | - Jerry Ware
- University of Arkansas for Medical Sciences, Little Rock
| | - Baidong Hou
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Beijing, China
| | - Alex F de Vos
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Center (UMC), University of Amsterdam, The Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Center (UMC), University of Amsterdam, The Netherlands.,Division of Infectious Diseases, Academic Medical Center, Amsterdam UMC, University of Amsterdam, The Netherlands
| | - Cornelis van 't Veer
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Center (UMC), University of Amsterdam, The Netherlands
| | - W Joost Wiersinga
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Center (UMC), University of Amsterdam, The Netherlands.,Division of Infectious Diseases, Academic Medical Center, Amsterdam UMC, University of Amsterdam, The Netherlands
| |
Collapse
|
94
|
Beristain-Covarrubias N, Perez-Toledo M, Thomas MR, Henderson IR, Watson SP, Cunningham AF. Understanding Infection-Induced Thrombosis: Lessons Learned From Animal Models. Front Immunol 2019; 10:2569. [PMID: 31749809 PMCID: PMC6848062 DOI: 10.3389/fimmu.2019.02569] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/16/2019] [Indexed: 12/25/2022] Open
Abstract
Thrombosis is a common consequence of infection that is associated with poor patient outcome. Nevertheless, the mechanisms by which infection-associated thrombosis is induced, maintained and resolved are poorly understood, as is the contribution thrombosis makes to host control of infection and pathogen spread. The key difference between infection-associated thrombosis and thrombosis in other circumstances is a stronger inflammation-mediated component caused by the presence of the pathogen and its products. This inflammation triggers the activation of platelets, which may accompany damage to the endothelium, resulting in fibrin deposition and thrombus formation. This process is often referred to as thrombo-inflammation. Strikingly, despite its clinical importance and despite thrombi being induced to many different pathogens, it is still unclear whether the mechanisms underlying this process are conserved and how we can best understand this process. This review summarizes thrombosis in a variety of models, including single antigen models such as LPS, and infection models using viruses and bacteria. We provide a specific focus on Salmonella Typhimurium infection as a useful model to address all stages of thrombosis during infection. We highlight how this model has helped us identify how thrombosis can appear in different organs at different times and thrombi be detected for weeks after infection in one site, yet largely be resolved within 24 h in another. Furthermore, we discuss the observation that thrombi induced to Salmonella Typhimurium are largely devoid of bacteria. Finally, we discuss the value of different therapeutic approaches to target thrombosis, the potential importance of timing in their administration and the necessity to maintain normal hemostasis after treatment. Improvements in our understanding of these processes can be used to better target infection-mediated mechanisms of thrombosis.
Collapse
Affiliation(s)
- Nonantzin Beristain-Covarrubias
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Marisol Perez-Toledo
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Mark R Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Ian R Henderson
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Midlands, United Kingdom
| | - Adam F Cunningham
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
95
|
Zindel J, Kubes P. DAMPs, PAMPs, and LAMPs in Immunity and Sterile Inflammation. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2019; 15:493-518. [PMID: 31675482 DOI: 10.1146/annurev-pathmechdis-012419-032847] [Citation(s) in RCA: 431] [Impact Index Per Article: 86.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recognizing the importance of leukocyte trafficking in inflammation led to some therapeutic breakthroughs. However, many inflammatory pathologies remain without specific therapy. This review discusses leukocytes in the context of sterile inflammation, a process caused by sterile (non-microbial) molecules, comprising damage-associated molecular patterns (DAMPs). DAMPs bind specific receptors to activate inflammation and start a highly optimized sequence of immune cell recruitment of neutrophils and monocytes to initiate effective tissue repair. When DAMPs are cleared, the recruited leukocytes change from a proinflammatory to a reparative program, a switch that is locally supervised by invariant natural killer T cells. In addition, neutrophils exit the inflammatory site and reverse transmigrate back to the bloodstream. Inflammation persists when the program switch or reverse transmigration fails, or when the coordinated leukocyte effort cannot clear the immunostimulatory molecules. The latter causes inappropriate leukocyte activation, a driver of many pathologies associated with poor lifestyle choices. We discuss lifestyle-associated inflammatory diseases and their corresponding immunostimulatory lifestyle-associated molecular patterns (LAMPs) and distinguish them from DAMPs.
Collapse
Affiliation(s)
- Joel Zindel
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N 4N1, Canada; .,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Department of Visceral Surgery and Medicine, Department for BioMedical Research, University of Bern, CH-3008 Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, CH-3012 Bern, Switzerland
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta T2N 4N1, Canada; .,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada.,Department of Microbiology, Immunology & Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
96
|
McDonald B, Dunbar M. Platelets and Intravascular Immunity: Guardians of the Vascular Space During Bloodstream Infections and Sepsis. Front Immunol 2019; 10:2400. [PMID: 31681291 PMCID: PMC6797619 DOI: 10.3389/fimmu.2019.02400] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 09/25/2019] [Indexed: 12/12/2022] Open
Abstract
Despite their humble origins as anuclear fragments of megakaryocytes, platelets have emerged as versatile mediators of thrombosis and immunity. The diverse spectrum of platelet functions are on full display during the host response to severe infection and sepsis, with platelets taking center-stage in the intravascular immune response to blood-borne pathogens. Platelets are endowed with a comprehensive armamentarium of pathogen detection systems that enable them to function as sentinels in the bloodstream for rapid identification of microbial invasion. Through both autonomous anti-microbial effector functions and collaborations with other innate immune cells, platelets orchestrate a complex intravascular immune defense system that protects against bacterial dissemination. As with any powerful immune defense system, dysregulation of platelet-mediated intravascular immunity can lead to profound collateral damage to host cells and tissues, resulting in sepsis-associated organ dysfunction. In this article, the cellular and molecular contributions of platelets to intravascular immune defenses in sepsis will be reviewed, including the roles of platelets in surveillance of the microcirculation and elicitation of protective anti-bacterial responses. Mechanisms of platelet-mediated thromboinflammatory organ dysfunction will be explored, with linkages to clinical biomarkers of platelet homeostasis that aid in the diagnosis and prognostication of human sepsis. Lastly, we discuss novel therapeutic opportunities that take advantage of our evolving understanding of platelets and intravascular immunity in severe infection.
Collapse
Affiliation(s)
- Braedon McDonald
- Department of Critical Care Medicine, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mary Dunbar
- Department of Pediatrics, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
97
|
Yadav VK, Singh PK, Agarwal V, Singh SK. Crosstalk between Platelet and Bacteria: A Therapeutic Prospect. Curr Pharm Des 2019; 25:4041-4052. [PMID: 31553286 DOI: 10.2174/1381612825666190925163347] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/13/2019] [Indexed: 12/16/2022]
Abstract
Platelets are typically recognized for their roles in the maintenance of hemostasis and vascular wall repair to reduce blood loss. Beyond hemostasis, platelets also play a critical role in pathophysiological conditions like atherosclerosis, stroke, thrombosis, and infections. During infection, platelets interact directly and indirectly with bacteria through a wide range of cellular and molecular mechanisms. Platelet surface receptors such as GPIbα, FcγRIIA, GPIIbIIIa, and TLRs, etc. facilitate direct interaction with bacterial cells. Besides, the indirect interaction between platelet and bacteria involves host plasma proteins such as von Willebrand Factor (vWF), fibronectin, IgG, and fibrinogen. Bacterial cells induce platelet activation, aggregation, and thrombus formation in the microvasculature. The activated platelets induce the Neutrophil Extracellular Traps (NETs) formation, which further contribute to thrombosis. Thus, platelets are extensively anticipated as vital immune modulator cells during infection, which may further lead to cardiovascular complications. In this review, we cover the interaction mechanisms between platelets and bacteria that may lead to the development of thrombotic disorders. Platelet receptors and other host molecules involved in such interactions can be used to develop new therapeutic strategies to combat against infection-induced cardiovascular complications. In addition, we highlight other receptor and enzyme targets that may further reduce infection-induced platelet activation and various pathological conditions.
Collapse
Affiliation(s)
- Vivek K Yadav
- Department of Biotechnology Motilal Nehru National Institute of Technology Allahabad, Prayagraj, India
| | - Pradeep K Singh
- Department of Biotechnology Motilal Nehru National Institute of Technology Allahabad, Prayagraj, India
| | - Vishnu Agarwal
- Department of Biotechnology Motilal Nehru National Institute of Technology Allahabad, Prayagraj, India
| | - Sunil K Singh
- Department of Animal Sciences, Central University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
98
|
Guo L, Rondina MT. The Era of Thromboinflammation: Platelets Are Dynamic Sensors and Effector Cells During Infectious Diseases. Front Immunol 2019; 10:2204. [PMID: 31572400 PMCID: PMC6753373 DOI: 10.3389/fimmu.2019.02204] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/30/2019] [Indexed: 12/12/2022] Open
Abstract
Platelets are anucleate cells produced by megakaryocytes. In recent years, a robust body of literature supports the evolving role of platelets as key sentinel and effector cells in infectious diseases, especially critical in bridging hemostatic, inflammatory, and immune continuums. Upon intravascular pathogen invasion, platelets can directly sense viral, parasitic, and bacterial infections through pattern recognition receptors and integrin receptors or pathogen: immunoglobulin complexes through Fc and complement receptors—although our understanding of these interactions remains incomplete. Constantly scanning for areas of injury or inflammation as they circulate in the vasculature, platelets also indirectly respond to pathogen invasion through interactions with leukocytes and the endothelium. Following antigen recognition, platelets often become activated. Through a diverse repertoire of mechanisms, activated platelets can directly sequester or kill pathogens, or facilitate pathogen clearance by activating macrophages and neutrophils, promoting neutrophil extracellular traps (NETs) formation, forming platelet aggregates and microthrombi. At times, however, platelet activation may also be injurious to the host, exacerbating inflammation and promoting endothelial damage and thrombosis. There are many gaps in our understandings of the role of platelets in infectious diseases. However, with the emergence of advanced technologies, our knowledge is increasing. In the current review, we mainly discuss these evolving roles of platelets under four different infectious pathogen infections, of which are dengue, malaria, Esterichia coli (E. coli) and staphylococcus aureus S. aureus, highlighting the complex interplay of these processes with hemostatic and thrombotic pathways.
Collapse
Affiliation(s)
- Li Guo
- University of Utah Molecular Medicine Program, Salt Lake City, UT, United States
| | - Matthew T Rondina
- University of Utah Molecular Medicine Program, Salt Lake City, UT, United States.,Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States.,Department of Pathology, University of Utah, Salt Lake City, UT, United States.,George E. Wahlen VAMC Department of Internal Medicine and GRECC, Salt Lake City, UT, United States
| |
Collapse
|
99
|
Abstract
Acute kidney injury (AKI), a major public health problem associated with high mortality and increased risk of progression towards end-stage renal disease, is characterized by the activation of intra-renal haemostatic and inflammatory processes. Platelets, which are present in high numbers in the circulation and can rapidly release a broad spectrum of bioactive mediators, are important acute modulators of inflammation and haemostasis, as they are the first cells to arrive at sites of acute injury, where they interact with endothelial cells and leukocytes. Diminished control of platelet reactivity by endothelial cells and/or an increased release of platelet-activating mediators can lead to uncontrolled platelet activation in AKI. As increased platelet sequestration and increased expression levels of the markers P-selectin, thromboxane A2, CC-chemokine ligand 5 and platelet factor 4 on platelets have been reported in kidneys following AKI, platelet activation likely plays a part in AKI pathology. Results from animal models and some clinical studies highlight the potential of antiplatelet therapies in the preservation of renal function in the context of AKI, but as current strategies also affect other cell types and non-platelet-derived mediators, additional studies are required to further elucidate the extent of platelet contribution to the pathology of AKI and to determine the best therapeutic approach by which to specifically target related pathogenic pathways.
Collapse
Affiliation(s)
- Marcel P B Jansen
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Sandrine Florquin
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
100
|
Assinger A, Schrottmaier WC, Salzmann M, Rayes J. Platelets in Sepsis: An Update on Experimental Models and Clinical Data. Front Immunol 2019; 10:1687. [PMID: 31379873 PMCID: PMC6650595 DOI: 10.3389/fimmu.2019.01687] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/04/2019] [Indexed: 12/22/2022] Open
Abstract
Beyond their important role in hemostasis, platelets play a crucial role in inflammatory diseases. This becomes apparent during sepsis, where platelet count and activation correlate with disease outcome and survival. Sepsis is caused by a dysregulated host response to infection, leading to organ dysfunction, permanent disabilities, or death. During sepsis, tissue injury results from the concomitant uncontrolled activation of the complement, coagulation, and inflammatory systems as well as platelet dysfunction. The balance between the systemic inflammatory response syndrome (SIRS) and the compensatory anti-inflammatory response (CARS) regulates sepsis outcome. Persistent thrombocytopenia is considered as an independent risk factor of mortality in sepsis, although it is still unclear whether the drop in platelet count is the cause or the consequence of sepsis severity. The role of platelets in sepsis development and progression was addressed in different experimental in vivo models, particularly in mice, that represent various aspects of human sepsis. The immunomodulatory function of platelets depends on the experimental model, time, and type of infection. Understanding the molecular mechanism of platelet regulation in inflammation could bring us one step closer to understand this important aspect of primary hemostasis which drives thrombotic as well as bleeding complications in patients with sterile and infectious inflammation. In this review, we summarize the current understanding of the contribution of platelets to sepsis severity and outcome. We highlight the differences between platelet receptors in mice and humans and discuss the potential and limitations of animal models to study platelet-related functions in sepsis.
Collapse
Affiliation(s)
- Alice Assinger
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | - Manuel Salzmann
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Julie Rayes
- Institute of Cardiovascular Science, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|