1
|
Starikova EA, Mammedova JT, Rubinstein AA, Sokolov AV, Kudryavtsev IV. Activation of the Coagulation Cascade as a Universal Danger Sign. Curr Issues Mol Biol 2025; 47:108. [PMID: 39996829 PMCID: PMC11854423 DOI: 10.3390/cimb47020108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/29/2025] [Accepted: 02/02/2025] [Indexed: 02/26/2025] Open
Abstract
Hemostasis is a mechanism that stops bleeding from an injured vessel, involves multiple interlinked steps, culminating in the formation of a "clot" sealing the damaged area. Moreover, it has long been recognized that inflammation also provokes the activation of the coagulation system. However, there has been an increasing amount of evidence revealing the immune function of the hemostasis system. This review collects and analyzes the results of the experimental studies and data from clinical observations confirming the inflammatory function of hemostasis. Here, we summarize the latest knowledge of the pathways in immune system activation under the influence of coagulation factors. The data analyzed allow us to consider the components of hemostasis as receptors recognizing «foreign» or damaged «self» or/and as «self» damage signals that initiate and reinforce inflammation and affect the direction of the adaptive immune response. To sum up, the findings collected in the review allow us to classify the coagulation factors, such as Damage-Associated Molecular Patterns that break down the conventional concepts of the coagulation system.
Collapse
Affiliation(s)
- Eleonora A. Starikova
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
- Department of Microbiology and Virology, Institute of Medical Education Almazov National Medical Research Centre, 2 Akkuratova Street, 197341 Saint Petersburg, Russia
| | - Jennet T. Mammedova
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
- Department of Molecular Biotechnology, Chemical and Biotechnology Faculty, Saint Petersburg State Institute of Technology, Moskovski Ave., 26, 190013 Saint Petersburg, Russia
| | - Artem A. Rubinstein
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
| | - Alexey V. Sokolov
- Laboratory of Systemic Virology, Department of Molecular Biology of Viruses, Smorodintsev Research Institute of Influenza, 15/17, Prof. Popova Str., 197376 Saint Petersburg, Russia;
| | - Igor V. Kudryavtsev
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| |
Collapse
|
2
|
Peshkova AD, Saliakhutdinova SM, Sounbuli K, Selivanova YA, Andrianova IA, Khabirova AI, Litvinov RI, Weisel JW. The differential formation and composition of leukocyte-platelet aggregates induced by various cellular stimulants. Thromb Res 2024; 241:109092. [PMID: 39024901 PMCID: PMC11411814 DOI: 10.1016/j.thromres.2024.109092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/16/2024] [Accepted: 07/05/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Leukocyte-platelet aggregates comprise a pathogenic link between hemostasis and immunity, but the prerequisites and mechanisms of their formation remain not understood. AIMS To quantify the formation, composition, and morphology of leukocyte-platelet aggregates in vitro under the influence of various cellular activators. METHODS Phorbol-12-myristate-13-acetate (PMA), lipopolysaccharide (LPS), thrombin receptor-activating peptide (TRAP-6), and adenosine diphosphate (ADP) were used as cellular activators. Flow cytometry was utilized to identify and quantify aggregates in whole human blood and platelet-rich plasma. Cell types and cellular aggregates were identified using fluorescently labeled antibodies against the appropriate cellular markers, and cell activation was assessed by the expression of appropriate surface markers. For confocal fluorescent microscopy, cell membranes and nuclei were labeled. Neutrophil-platelet aggregates were studied using scanning electron microscopy. RESULTS In the presence of PMA, ADP or TRAP-6, about 17-38 % of neutrophils and 61-77 % of monocytes formed aggregates with platelets in whole blood, whereas LPS did not induce platelet aggregation with either neutrophils or monocytes due the inability to activate platelets. Similar results were obtained when isolated neutrophils were added to platelet-rich plasma. All the cell types involved in the heterotypic aggregation expressed molecular markers of activation. Fluorescent and electron microscopy of the aggregates showed that the predominant platelet/leukocyte ratios were 1:1 and 2:1. CONCLUSIONS Formation of leukocyte-platelet aggregates depends on the nature of the cellular activator and the spectrum of its cell-activating ability. An indispensable condition for formation of leukocyte-platelet aggregates is activation of all cell types including platelets, which is the restrictive step.
Collapse
Affiliation(s)
- Alina D Peshkova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | | | - Khetam Sounbuli
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Yuliya A Selivanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Izabella A Andrianova
- Department of Internal Medicine, Division of Hematology and Program in Molecular Medicine, University of Utah, Salt Lake City, UT, USA
| | - Alina I Khabirova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russian Federation
| | - Rustem I Litvinov
- Departments of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - John W Weisel
- Departments of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Duchez AC, Heestermans M, Arthaud CA, Eyraud MA, Portier M, Prier A, Hamzeh-Cognasse H, Cognasse F. In platelet single donor apheresis, platelet factor 4 levels correlated with donor's age and decreased during storage. Sci Rep 2024; 14:6231. [PMID: 38485973 PMCID: PMC10940288 DOI: 10.1038/s41598-024-56826-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/12/2024] [Indexed: 03/18/2024] Open
Abstract
The human population is ageing worldwide. The World Health Organization estimated that the world's population of people aged 60 years and older will increase to at least 30%, coinciding with a growing frequency of cognitive and cardiovascular disease. Recently, in preclinical studies platelet Factor 4 (PF4) was presented as a pro-cognitive factor. This molecule is released by platelets in the circulation and could be present in blood products destined for transfusion. We wondered if PF4 levels are correlated to the age of the blood donor or to the storage time of platelet concentrates (PCs) intended for transfusion? We observed higher levels of PF4 in PCs from elderly donors compared to younger donors, while PC storage time did not determine PF4 levels expression.
Collapse
Affiliation(s)
- Anne Claire Duchez
- INSERM, U 1059 SAINBIOSE, Université Jean Monnet, Mines Saint-Étienne, 42023, Saint-Etienne, France
- Establissement Français du Sang Auvergne-Rhône-Alpes and INSERM U1059, 25 Boulevard Pasteur, 42100, Saint-Etienne, France
| | - Marco Heestermans
- INSERM, U 1059 SAINBIOSE, Université Jean Monnet, Mines Saint-Étienne, 42023, Saint-Etienne, France
- Establissement Français du Sang Auvergne-Rhône-Alpes and INSERM U1059, 25 Boulevard Pasteur, 42100, Saint-Etienne, France
| | - Charles-Antoine Arthaud
- INSERM, U 1059 SAINBIOSE, Université Jean Monnet, Mines Saint-Étienne, 42023, Saint-Etienne, France
- Establissement Français du Sang Auvergne-Rhône-Alpes and INSERM U1059, 25 Boulevard Pasteur, 42100, Saint-Etienne, France
| | - Marie-Ange Eyraud
- INSERM, U 1059 SAINBIOSE, Université Jean Monnet, Mines Saint-Étienne, 42023, Saint-Etienne, France
- Establissement Français du Sang Auvergne-Rhône-Alpes and INSERM U1059, 25 Boulevard Pasteur, 42100, Saint-Etienne, France
| | - Mailys Portier
- INSERM, U 1059 SAINBIOSE, Université Jean Monnet, Mines Saint-Étienne, 42023, Saint-Etienne, France
- Establissement Français du Sang Auvergne-Rhône-Alpes and INSERM U1059, 25 Boulevard Pasteur, 42100, Saint-Etienne, France
| | - Amélie Prier
- INSERM, U 1059 SAINBIOSE, Université Jean Monnet, Mines Saint-Étienne, 42023, Saint-Etienne, France
- Establissement Français du Sang Auvergne-Rhône-Alpes and INSERM U1059, 25 Boulevard Pasteur, 42100, Saint-Etienne, France
| | - Hind Hamzeh-Cognasse
- INSERM, U 1059 SAINBIOSE, Université Jean Monnet, Mines Saint-Étienne, 42023, Saint-Etienne, France
| | - Fabrice Cognasse
- INSERM, U 1059 SAINBIOSE, Université Jean Monnet, Mines Saint-Étienne, 42023, Saint-Etienne, France.
- Establissement Français du Sang Auvergne-Rhône-Alpes and INSERM U1059, 25 Boulevard Pasteur, 42100, Saint-Etienne, France.
| |
Collapse
|
4
|
Baroni Pietto MC, Glembotsky AC, Lev PR, Marín Oyarzún CR, De Luca G, Gomez G, Collado MV, Charó N, Cellucci AS, Heller PG, Goette NP, Marta RF. Toll-like receptor expression and functional behavior in platelets from patients with systemic lupus erythematosus. Immunobiology 2024; 229:152782. [PMID: 38159527 DOI: 10.1016/j.imbio.2023.152782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/06/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Multiple blood cell abnormalities participate in the development of inflammation in systemic lupus erythematosus (SLE). Although platelets have been suggested as one of these contributors through the release of their content during activation, there are limited specific data about their role as immune players in SLE. MATERIALS AND METHODS Thirteen SLE patients were included. Flow cytometry was used to measure Toll-like receptors (TLR) 2, 4, and 9 in resting platelets, platelet-activation markers (PAC-1 binding, P-selectin, CD63, and CD40 ligand -L) and platelet-leukocyte aggregates before and after specific TLR stimulation. Soluble CD40L and von Willebrand factor (vWf) release from stimulated platelets was measured using ELISA. RESULTS In resting conditions, SLE platelets showed normal expression levels of TLR 2, 4 and 9. Platelet surface activation markers, soluble CD40L, and vWf release were normal at baseline and after TLR stimulation. Platelet-monocyte aggregates were elevated in resting conditions in SLE samples and showed only a marginal increase after TLR stimulation, while baseline and stimulated platelet-neutrophil and platelet-lymphocyte aggregates were normal. C-reactive protein levels positively correlated with platelet-monocyte aggregates both at baseline and after stimulation with the TLR-2 agonist PAM3CSK4, suggesting these complexes could reflect the inflammatory activity in SLE. In our cohort, 12 of 13 patients received treatment with hydroxychloroquine (HCQ), a known inhibitor of endosomal activity and a potential inhibitor of platelet activation. The fact that SLE platelets showed an adequate response to TLR agonists suggests that, despite this treatment, they retain the ability to respond to the increased levels of damage-associated molecular patterns (DAMPs), which represent known TLR ligands, present in the circulation of SLE patients. Interestingly, elevated plasma levels of high mobility group box 1 (HMGB1), a classical DAMP, correlated with vWf release from TLR-stimulated platelets, suggesting that HMGB1 may also be released by platelets, thereby creating a positive feedback loop for platelet activation that contributes to inflammation. CONCLUSION Our study demonstrates normal platelet TLR expression and function together with increased circulating platelet-monocyte aggregates. In addition, a direct correlation was observed between plasma HMGB1 levels and platelet vWf release following TLR2 stimulation. This platelet behavior in a group of patients undergoing HCQ treatment suggests that platelets could play a role in the inflammatory state of SLE.
Collapse
Affiliation(s)
- María C Baroni Pietto
- University of Buenos Aires, School of Medicine, Institute for Medical Research Alfredo Lanari. Buenos Aires, Argentina; National Scientific and Technical Research Council, University of Buenos Aires, Institute for Medical Research, Department of Hematology Research. Buenos Aires, Argentina
| | - Ana C Glembotsky
- University of Buenos Aires, School of Medicine, Institute for Medical Research Alfredo Lanari. Buenos Aires, Argentina; National Scientific and Technical Research Council, University of Buenos Aires, Institute for Medical Research, Department of Hematology Research. Buenos Aires, Argentina
| | - Paola R Lev
- University of Buenos Aires, School of Medicine, Institute for Medical Research Alfredo Lanari. Buenos Aires, Argentina; National Scientific and Technical Research Council, University of Buenos Aires, Institute for Medical Research, Department of Hematology Research. Buenos Aires, Argentina
| | - Cecilia R Marín Oyarzún
- University of Buenos Aires, School of Medicine, Institute for Medical Research Alfredo Lanari. Buenos Aires, Argentina; National Scientific and Technical Research Council, University of Buenos Aires, Institute for Medical Research, Department of Hematology Research. Buenos Aires, Argentina
| | - Geraldine De Luca
- University of Buenos Aires, School of Medicine, Institute for Medical Research Alfredo Lanari. Buenos Aires, Argentina; National Scientific and Technical Research Council, University of Buenos Aires, Institute for Medical Research, Department of Hematology Research. Buenos Aires, Argentina
| | - Graciela Gomez
- University of Buenos Aires, School of Medicine, Institute for Medical Research Alfredo Lanari. Department of Rheumatology. Buenos Aires, Argentina
| | - María V Collado
- University of Buenos Aires, School of Medicine, Institute for Medical Research Alfredo Lanari. Department of Rheumatology. Buenos Aires, Argentina
| | - Nancy Charó
- National Scientific and Technical Research Council-National Academy of Medicine, Institute of Experimental Medicine, Laboratory of Experimental Thrombosis and Immunobiology of Inflammation. Buenos Aires, Argentina
| | - Adela S Cellucci
- University of Buenos Aires, School of Medicine, Institute for Medical Research Alfredo Lanari. Buenos Aires, Argentina; National Scientific and Technical Research Council, University of Buenos Aires, Institute for Medical Research, Department of Hematology Research. Buenos Aires, Argentina
| | - Paula G Heller
- University of Buenos Aires, School of Medicine, Institute for Medical Research Alfredo Lanari. Buenos Aires, Argentina; National Scientific and Technical Research Council, University of Buenos Aires, Institute for Medical Research, Department of Hematology Research. Buenos Aires, Argentina
| | - Nora P Goette
- University of Buenos Aires, School of Medicine, Institute for Medical Research Alfredo Lanari. Buenos Aires, Argentina
| | - Rosana F Marta
- University of Buenos Aires, School of Medicine, Institute for Medical Research Alfredo Lanari. Buenos Aires, Argentina; National Scientific and Technical Research Council, University of Buenos Aires, Institute for Medical Research, Department of Hematology Research. Buenos Aires, Argentina.
| |
Collapse
|
5
|
Karmacharya M, Kumar S, Choi YJ, Cho YK. Platelet Membrane-Enclosed Bioorthogonal Catalysis for Combating Dental Caries. Adv Healthc Mater 2024; 13:e2302121. [PMID: 37847511 DOI: 10.1002/adhm.202302121] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/27/2023] [Indexed: 10/18/2023]
Abstract
Platelets have shown promise as a means to combat bacterial infections, fostering the development of innovative therapeutic approaches. However, several challenges persist, including cargo loading issues, limited efficacy against biofilms, and concerns regarding the impact of payloads on the platelet carriers. Here, human platelet membrane vesicles (h-PMVs) encapsulating supramolecular metal catalysts (SMCs) as "nanofactories" to convert prodrugs into antimicrobial compounds within close proximity to bacteria are introduced. Having established the feasibility and effectiveness of the SMCs within h-PMVs, referred to as the PLT-reactor, to activate pro-antibiotic drugs (pro-ciprofloxacin and pro-moxifloxacin) using model organisms (Escherichia coli ATCC 25922 and Staphylococcus aureus ATCC 25923), the investigation is subsequently extended to oral biofilms, with a particular emphasis on Streptococcus mutans 3065. This "bind and kill" strategy demonstrates the potent antimicrobial specificity of the PLT-reactor through localized antibiotic production. h-PMVs play a pivotal role by enabling precise targeting of pathogenic biofilms on natural teeth while minimizing potential hemolytic effects. The finding indicates that platelet membrane-cloaked surfaces exhibit robust, multifaceted, and pathogen-specific binding affinity with excellent biocompatibility, making them a promising alternative to antibody-based therapies for infectious diseases.
Collapse
Affiliation(s)
- Mamata Karmacharya
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, South Korea
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, South Korea
| | - Sumit Kumar
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, South Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, South Korea
| | - Yoon Jeong Choi
- Department of Orthodontics, Institute of Craniofacial Deformity, Yonsei University, College of Dentistry, Seoul, 03722, South Korea
| | - Yoon-Kyoung Cho
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan, 44919, South Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, South Korea
| |
Collapse
|
6
|
Khelfa M, Leclerc M, Kerbrat S, Boudjemai YNS, Benchouaia M, Neyrinck-Leglantier D, Cagnet L, Berradhia L, Tamagne M, Croisille L, Pirenne F, Maury S, Vingert B. Divergent CD4 + T-cell profiles are associated with anti-HLA alloimmunization status in platelet-transfused AML patients. Front Immunol 2023; 14:1165973. [PMID: 37701444 PMCID: PMC10493329 DOI: 10.3389/fimmu.2023.1165973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 07/17/2023] [Indexed: 09/14/2023] Open
Abstract
Introduction Acute myeloid leukemia (AML) is one of the commonest hematologic disorders. Due to the high frequency of disease- or treatment-related thrombocytopenia, AML requires treatment with multiple platelet transfusions, which can trigger a humoral response directed against platelets. Some, but not all, AML patients develop an anti-HLA immune response after multiple transfusions. We therefore hypothesized that different immune activation profiles might be associated with anti-HLA alloimmunization status. Methods We tested this hypothesis, by analyzing CD4+ T lymphocyte (TL) subsets and their immune control molecules in flow cytometry and single-cell multi-omics. Results A comparison of immunological status between anti-HLA alloimmunized and non-alloimmunized AML patients identified differences in the phenotype and function of CD4+ TLs. CD4+ TLs from alloimmunized patients displayed features of immune activation, with higher levels of CD40 and OX40 than the cells of healthy donors. However, the most notable differences were observed in non-alloimmunized patients. These patients had lower levels of CD40 and OX40 than alloimmunized patients and higher levels of PD1. Moreover, the Treg compartment of non-alloimmunized patients was larger and more functional than that in alloimmunized patients. These results were supported by a multi-omics analysis of immune response molecules in conventional CD4+ TLs, Tfh circulating cells, and Tregs. Discussion Our results thus reveal divergent CD4+ TL characteristics correlated with anti-HLA alloimmunization status in transfused AML patients. These differences, characterizing CD4+ TLs independently of any specific antigen, should be taken into account when considering the immune responses of patients to infections, vaccinations, or transplantations.
Collapse
Affiliation(s)
- Mehdi Khelfa
- Établissement Français du Sang, Île-de-France, France
- Univ Paris Est Creteil, INSERM, IMRB, Équipe Pirenne, Créteil, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Mathieu Leclerc
- Assistance Publique - Hôpitaux de Paris, Hôpital Henri Mondor, Service d’Hématologie clinique, Créteil, France
| | - Stéphane Kerbrat
- Univ Paris Est Creteil, INSERM, IMRB, Plateforme de Génomique, Créteil, France
| | | | - Médine Benchouaia
- Univ Paris Est Creteil, INSERM, IMRB, Plateforme de Génomique, Créteil, France
| | - Déborah Neyrinck-Leglantier
- Établissement Français du Sang, Île-de-France, France
- Univ Paris Est Creteil, INSERM, IMRB, Équipe Pirenne, Créteil, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Léonie Cagnet
- Établissement Français du Sang, Île-de-France, France
- Univ Paris Est Creteil, INSERM, IMRB, Équipe Pirenne, Créteil, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Lylia Berradhia
- Établissement Français du Sang, Île-de-France, France
- Univ Paris Est Creteil, INSERM, IMRB, Équipe Pirenne, Créteil, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Marie Tamagne
- Établissement Français du Sang, Île-de-France, France
- Univ Paris Est Creteil, INSERM, IMRB, Équipe Pirenne, Créteil, France
- Laboratory of Excellence GR-Ex, Paris, France
| | | | - France Pirenne
- Établissement Français du Sang, Île-de-France, France
- Univ Paris Est Creteil, INSERM, IMRB, Équipe Pirenne, Créteil, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Sébastien Maury
- Assistance Publique - Hôpitaux de Paris, Hôpital Henri Mondor, Service d’Hématologie clinique, Créteil, France
| | - Benoît Vingert
- Établissement Français du Sang, Île-de-France, France
- Univ Paris Est Creteil, INSERM, IMRB, Équipe Pirenne, Créteil, France
- Laboratory of Excellence GR-Ex, Paris, France
| |
Collapse
|
7
|
El Hasbani G, Saliba AN, Uthman I, Taher AT. Hematological manifestations of antiphospholipid syndrome: Going beyond thrombosis. Blood Rev 2023; 58:101015. [PMID: 36175215 DOI: 10.1016/j.blre.2022.101015] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022]
Abstract
Thrombotic complications are a hallmark of antiphospholipid syndrome (APS). These vascular - arterial, venous, and/or small vessel - complications are well described and known to hematologists and healthcare providers caring for patients with this disease. In this review, we shed light on other hematological manifestations of the disease, including bleeding, thrombocytopenia, autoimmune hemolytic anemia, and thrombotic microangiopathy syndromes. While these manifestations are not bona fide clinical criteria for the diagnosis of APS, they frequently interact and contribute to the complexity of clinical management of APS.
Collapse
Affiliation(s)
- Georges El Hasbani
- Department of Internal Medicine, Hartford Healthcare, St. Vincent's Medical Center, Bridgeport, CT 06606, USA
| | - Antoine N Saliba
- Division of Hematology, Department of Medicine, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Imad Uthman
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali T Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon..
| |
Collapse
|
8
|
Tang X, Xu Q, Yang S, Huang X, Wang L, Huang F, Luo J, Zhou X, Wu A, Mei Q, Zhao C, Wu J. Toll-like Receptors and Thrombopoiesis. Int J Mol Sci 2023; 24:1010. [PMID: 36674552 PMCID: PMC9864288 DOI: 10.3390/ijms24021010] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 12/27/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Platelets are the second most abundant blood component after red blood cells and can participate in a variety of physiological and pathological functions. Beyond its traditional role in hemostasis and thrombosis, it also plays an indispensable role in inflammatory diseases. However, thrombocytopenia is a common hematologic problem in the clinic, and it presents a proportional relationship with the fatality of many diseases. Therefore, the prevention and treatment of thrombocytopenia is of great importance. The expression of Toll-like receptors (TLRs) is one of the most relevant characteristics of thrombopoiesis and the platelet inflammatory function. We know that the TLR family is found on the surface or inside almost all cells, where they perform many immune functions. Of those, TLR2 and TLR4 are the main stress-inducing members and play an integral role in inflammatory diseases and platelet production and function. Therefore, the aim of this review is to present and discuss the relationship between platelets, inflammation and the TLR family and extend recent research on the influence of the TLR2 and TLR4 pathways and the regulation of platelet production and function. Reviewing the interaction between TLRs and platelets in inflammation may be a research direction or program for the treatment of thrombocytopenia-related and inflammatory-related diseases.
Collapse
Affiliation(s)
- Xiaoqin Tang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Qian Xu
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Shuo Yang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Xinwu Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Long Wang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Feihong Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Jiesi Luo
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Xiaogang Zhou
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Anguo Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Qibing Mei
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Chunling Zhao
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Jianming Wu
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| |
Collapse
|
9
|
The Controversial Role of LPS in Platelet Activation In Vitro. Int J Mol Sci 2022; 23:ijms231810900. [PMID: 36142813 PMCID: PMC9505944 DOI: 10.3390/ijms231810900] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Circulating platelets are responsible for hemostasis and thrombosis but are also primary sensors of pathogens and are involved in innate immunity, inflammation, and sepsis. Sepsis is commonly caused by an exaggerated immune response to bacterial, viral, and fungal infections, and leads to severe thrombotic complications. Among others, the endotoxin lipopolysaccharide (LPS) found in the outer membrane of Gram-negative bacteria is the most common trigger of sepsis. Since the discovery of the expression of the LPS receptor TLR4 in platelets, several studies have investigated the ability of LPS to induce platelet activation and to contribute to a prothrombotic phenotype, per se or in combination with plasma proteins and platelet agonists. This issue, however, is still controversial, as different sources, purity, and concentrations of LPS, different platelet-purification protocols, and different methods of analysis have been used in the past two decades, giving contradictory results. This review summarizes and critically analyzes past and recent publications about LPS-induced platelet activation in vitro. A methodological section illustrates the principal platelet preparation protocols and significant differences. The ability of various sources of LPS to elicit platelet activation in terms of aggregation, granule secretion, cytokine release, ROS production, and interaction with leukocytes and NET formation is discussed.
Collapse
|
10
|
de Oliveira AA, Mendoza VO, Rastogi S, Nunes KP. New insights into the role and therapeutic potential of HSP70 in diabetes. Pharmacol Res 2022; 178:106173. [PMID: 35278625 DOI: 10.1016/j.phrs.2022.106173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/06/2022] [Accepted: 03/07/2022] [Indexed: 10/18/2022]
Abstract
Emerging evidence indicates that HSP70 represents a key mechanism in the pathophysiology of β-cell dysfunction, insulin resistance, and various diabetic complications, including micro- and macro-vascular alterations, as well as impaired hemostasis. Hyperglycemia, a hallmark of both types of diabetes, increases the circulating levels of HSP70 (eHSP70), but there is still divergence about whether diabetes up- or down-regulates the intracellular fraction of this protein (iHSP70). Here, we consider that iHSP70 levels reduce in diabetic arterial structures and that the vascular system is in direct contact with all other systems in the body suggesting that a systemic response might also be happening for iHSP70, which is characterized by decreased levels of HSP70 in the vasculature. Furthermore, although many pathways have been proposed to explain HSP70's functions in diabetes, and organs/tissues/cells-specific variations occur, the membrane-bound receptor of the innate immune system, Toll-like receptor 4, and its downstream signal transduction pathways appear to be a constant, not only when we explore the actions of eHSP70, but also when we assess the contributions of iHSP70. In this review, we focus on discussing the multiple roles of HSP70 across organs/tissues/cells affected by hyperglycemia to further explore the possibility of targeting this protein with pharmacological and non-pharmacological approaches in the context of diabetes.
Collapse
Affiliation(s)
- Amanda Almeida de Oliveira
- Laboratory of Vascular Biology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, United States
| | - Valentina Ochoa Mendoza
- Laboratory of Vascular Biology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, United States
| | - Swasti Rastogi
- Laboratory of Vascular Biology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, United States
| | - Kenia Pedrosa Nunes
- Laboratory of Vascular Biology, Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, United States.
| |
Collapse
|
11
|
Serum Calcium Level Combined with Platelet Count May Be Useful Indicators for Assisted Diagnosis of Extremity Posttraumatic Osteomyelitis: A Comparative Analysis. DISEASE MARKERS 2021; 2021:6196663. [PMID: 34745387 PMCID: PMC8568509 DOI: 10.1155/2021/6196663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 09/01/2021] [Accepted: 10/16/2021] [Indexed: 11/17/2022]
Abstract
Background A previous study had reported that patients with osteomyelitis (OM) appeared to be more likely to develop hypocalcemia before and after surgery. Calcium sulfate (CS) is frequently used as a local antibiotic vehicle in the treatment of OM, which may also affect serum calcium level. However, whether changes of serum calcium level are caused by OM and/or local use of calcium sulfate remains unclear. Also, platelet (PLT) count plays a crucial predictive role in periprosthetic joint infections (PJIs), but its role in assisted diagnosis of OM is largely unknown. The purpose of this study was to determine whether serum calcium level and PLT count may be helpful in assisted diagnosis of PTOM. Methods Between January 2013 and December 2018, we analyzed 468 consecutive patients (392 males and 76 females), including 170 patients with posttraumatic OM (PTOM), 130 patients with aseptic bone nonunion (ABN), and 168 patients recovered from fractures with requirement of implant removal set as controls. Preoperative serological levels of calcium, phosphorus, and PLT were detected, and comparisons were conducted among the above three groups. Additionally, correlations and receiver operating characteristic (ROC) curves were displayed to test whether calcium level and PLT can differentiate patients with ABN and PTOM. Results Outcomes showed that the incidences of asymptomatic hypocalcemia (PTOM vs. ABN vs. controls = 22.94% vs. 6.92% vs. 8.82%, χ2 = 21.098, P < 0.001) and thrombocytosis (PTOM vs. ABN vs. controls = 35.3% vs. 13.84% vs. 12.35%, χ2 = 28.512, P < 0.001) were highest in PTOM patients. Besides, the mean serological levels of phosphorus in PTOM and ABN patients were significantly higher than those in the controls (P = 0.007). The Area Under the Curve (AUC) of the ROC curve outcomes revealed that, with the combination of serum calcium level with PLT count, the predictive role was acceptable (AUC 0.730, P < 0.001, 95% CI 0.681-0.780). Also, serological levels of calcium of 2.225 mmol/L and PLT count of 246.5 × 109/L were identified as the optimal cut-off values to distinguish patients with and without PTOM. However, age- and gender-related differences in serum calcium levels (age, P = 0.056; gender, P = 0.978) and PLT count (age, P = 0.363; gender, P = 0.799) were not found to be statistically significant in any groups. In addition, no significant correlations were identified between serum calcium level and PLT count (R = 0.010, P = 0.839). Conclusions Asymptomatic hypocalcemia and thrombocytosis appeared to be more frequent in this cohort with PTOM. Serological levels of calcium and PLT count may be useful biomarkers in screening patients suspected of PTOM.
Collapse
|
12
|
Page MJ, Pretorius E. Platelet Behavior Contributes to Neuropathologies: A Focus on Alzheimer's and Parkinson's Disease. Semin Thromb Hemost 2021; 48:382-404. [PMID: 34624913 DOI: 10.1055/s-0041-1733960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The functions of platelets are broad. Platelets function in hemostasis and thrombosis, inflammation and immune responses, vascular regulation, and host defense against invading pathogens, among others. These actions are achieved through the release of a wide set of coagulative, vascular, inflammatory, and other factors as well as diverse cell surface receptors involved in the same activities. As active participants in these physiological processes, platelets become involved in signaling pathways and pathological reactions that contribute to diseases that are defined by inflammation (including by pathogen-derived stimuli), vascular dysfunction, and coagulation. These diseases include Alzheimer's and Parkinson's disease, the two most common neurodegenerative diseases. Despite their unique pathological and clinical features, significant shared pathological processes exist between these two conditions, particularly relating to a central inflammatory mechanism involving both neuroinflammation and inflammation in the systemic environment, but also neurovascular dysfunction and coagulopathy, processes which also share initiation factors and receptors. This triad of dysfunction-(neuro)inflammation, neurovascular dysfunction, and hypercoagulation-illustrates the important roles platelets play in neuropathology. Although some mechanisms are understudied in Alzheimer's and Parkinson's disease, a strong case can be made for the relevance of platelets in neurodegeneration-related processes.
Collapse
Affiliation(s)
- Martin J Page
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, South Africa
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, South Africa
| |
Collapse
|
13
|
Wilson-Nieuwenhuis J, El-Mohtadi M, Edwards K, Whitehead K, Dempsey-Hibbert N. Factors Involved in the onset of infection following bacterially contaminated platelet transfusions. Platelets 2021; 32:909-918. [PMID: 32762589 DOI: 10.1080/09537104.2020.1803253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Transfusion of platelet concentrates (PCs) is associated with several adverse patient reactions, the most common of which are febrile non-hemolytic transfusion reactions (FNHTRs) and transfusion-associated bacterial-infection/transfusion-associated sepsis (T-ABI/TA-S). Diagnosis of T-ABI/T-AS requires a positive blood culture (BC) result from the transfusion recipient and also a positive identification of bacterial contamination within a test aliquot of the transfused PC. In a significant number of cases, clinical symptoms post-transfusion are reported by the clinician, yet the BCs from the patient and/or PC are negative. The topic of 'missed bacterial detection' has therefore been the focus of several primary research studies and review articles, suggesting that biofilm formation in the blood bag and the presence of viable but non-culturable (VBNC) pathogens are the major causes of this missed detection. However, platelets are emerging as key players in early host responses to infection and as such, the aforementioned biofilm formation could elicit 'platelet priming', which could lead to significant immunological reactions in the host, in the absence of planktonic bacteria in the host bloodstream. This review reflects on what is known about missed detection and relates this to the emerging understanding of the effect of bacterial contamination on the platelets themselves and the significant role played by platelets in exacerbation of an immune response to infection within the transfusion setting.
Collapse
Affiliation(s)
| | - Mohamed El-Mohtadi
- Centre for Bioscience, Manchester Metropolitan University, Manchester, UK
| | - Kurtis Edwards
- Centre for Bioscience, Manchester Metropolitan University, Manchester, UK
| | - Kathryn Whitehead
- Centre for Bioscience, Manchester Metropolitan University, Manchester, UK
| | | |
Collapse
|
14
|
Rolfes V, Ribeiro LS, Hawwari I, Böttcher L, Rosero N, Maasewerd S, Santos MLS, Próchnicki T, Silva CMDS, Wanderley CWDS, Rothe M, Schmidt SV, Stunden HJ, Bertheloot D, Rivas MN, Fontes CJ, Carvalho LH, Cunha FQ, Latz E, Arditi M, Franklin BS. Platelets Fuel the Inflammasome Activation of Innate Immune Cells. Cell Rep 2021; 31:107615. [PMID: 32402278 PMCID: PMC7225754 DOI: 10.1016/j.celrep.2020.107615] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 03/12/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
The inflammasomes control the bioactivity of pro-inflammatory cytokines of the interleukin (IL)-1 family. The inflammasome assembled by NLRP3 has been predominantly studied in homogeneous cell populations in vitro, neglecting the influence of cellular interactions that occur in vivo. Here, we show that platelets boost the inflammasome capacity of human macrophages and neutrophils and are critical for IL-1 production by monocytes. Platelets license NLRP3 transcription, thereby enhancing ASC oligomerization, caspase-1 activity, and IL-1β secretion. Platelets influence IL-1β production in vivo, and blood platelet counts correlate with plasmatic IL-1β levels in malaria. Furthermore, we reveal an enriched platelet gene signature among the highest-expressed transcripts in IL-1β-driven autoinflammatory diseases. The platelet effect is independent of cell-to-cell contact, platelet-derived lipid mediators, purines, nucleic acids, and a host of platelet cytokines, and it involves the triggering of calcium-sensing receptors on macrophages. Hence, platelets provide an additional layer of regulation of inflammasomes and IL-1-driven inflammation. Platelets license NLRP3 for inflammasome activattion in innate immune cells Platelets are required for optimal monocyte inflammasome activation Platelets shape IL-1β in vivo, and platelet counts correlate with IL-1β in plasma A constitutive, heat-sensitive soluble platelet-factor boost IL-1β in macrophages
Collapse
Affiliation(s)
- Verena Rolfes
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Lucas Secchim Ribeiro
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany.
| | - Ibrahim Hawwari
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Lisa Böttcher
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Nathalia Rosero
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Salie Maasewerd
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Marina Lima Silva Santos
- Laboratório de Malária, Instituto René Rachou, Fundação Oswaldo Cruz, 30190-002 Belo Horizonte, MG, Brazil
| | - Tomasz Próchnicki
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Camila Meirelles de Souza Silva
- Center for Research in Inflammatory Diseases, School of Medicine of Ribeirão Preto, University of Sao Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Carlos Wagner de Souza Wanderley
- Center for Research in Inflammatory Diseases, School of Medicine of Ribeirão Preto, University of Sao Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Maximilian Rothe
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Susanne V Schmidt
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - H James Stunden
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Damien Bertheloot
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Magali Noval Rivas
- Departments of Pediatrics, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Cor Jesus Fontes
- Departamento de Clínica Médica, Universidade Federal de Mato Grosso, 78060-900 Cuiabá, MT, Brazil
| | - Luzia Helena Carvalho
- Laboratório de Malária, Instituto René Rachou, Fundação Oswaldo Cruz, 30190-002 Belo Horizonte, MG, Brazil
| | - Fernando Queiroz Cunha
- Center for Research in Inflammatory Diseases, School of Medicine of Ribeirão Preto, University of Sao Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Eicke Latz
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany; Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01655, USA; German Center for Neurodegenerative Diseases, 53127 Bonn, NRW, Germany
| | - Moshe Arditi
- Departments of Pediatrics, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA.
| | | |
Collapse
|
15
|
Vulliamy P, Kornblith LZ, Kutcher ME, Cohen MJ, Brohi K, Neal MD. Alterations in platelet behavior after major trauma: adaptive or maladaptive? Platelets 2021; 32:295-304. [PMID: 31986948 PMCID: PMC7382983 DOI: 10.1080/09537104.2020.1718633] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/01/2020] [Accepted: 01/14/2020] [Indexed: 12/14/2022]
Abstract
Platelets are damage sentinels of the intravascular compartment, initiating and coordinating the primary response to tissue injury. Severe trauma and hemorrhage induce profound alterations in platelet behavior. During the acute post-injury phase, platelets develop a state of impaired ex vivo agonist responsiveness independent of platelet count, associated with systemic coagulopathy and mortality risk. In patients surviving the initial insult, platelets become hyper-responsive, associated with increased risk of thrombotic events. Beyond coagulation, platelets constitute part of a sterile inflammatory response to injury: both directly through release of immunomodulatory molecules, and indirectly through modifying behavior of innate leukocytes. Both procoagulant and proinflammatory aspects have implications for secondary organ injury and multiple-organ dysfunction syndromes. This review details our current understanding of adaptive and maladaptive alterations in platelet biology induced by severe trauma, mechanisms underlying these alterations, potential platelet-focused therapies, and existing knowledge gaps and their research implications.
Collapse
Affiliation(s)
- Paul Vulliamy
- Centre for Trauma Sciences, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, E1 2AT, United Kingdom
| | - Lucy Z. Kornblith
- Department of Surgery, Zuckerberg San Francisco General Hospital and the University of California, San Francisco, San Francisco, California
| | - Matthew E. Kutcher
- Division of Trauma, Critical Care, and Acute Care Surgery, University of Mississippi Medical Center, Jackson, Mississippi
| | - Mitchell J. Cohen
- Department of Surgery, University of Colorado, Aurora, Colorado
- Ernest E Moore Shock Trauma Center at Denver Health, Denver, Colorado
| | - Karim Brohi
- Centre for Trauma Sciences, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, E1 2AT, United Kingdom
| | - Matthew D. Neal
- Division of Trauma and Acute Care Surgery, Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
16
|
McMullen PD, Cho JH, Miller JL, Husain AN, Pytel P, Krausz T. A Descriptive and Quantitative Immunohistochemical Study Demonstrating a Spectrum of Platelet Recruitment Patterns Across Pulmonary Infections Including COVID-19. Am J Clin Pathol 2021; 155:354-363. [PMID: 33174599 PMCID: PMC7717231 DOI: 10.1093/ajcp/aqaa230] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Pulmonary platelet deposition and microangiopathy are increasingly recognized components of coronavirus disease 2019 (COVID-19) infection. Thrombosis is a known component of sepsis and disseminated intravascular coagulation. We sought to compare the level of platelet deposition in the pulmonary vasculature in cases of confirmed COVID-19 infection to other lung injuries and infections. METHODS Immunohistochemistry was performed on 27 autopsy cases and 2 surgical pathology cases targeting CD61. Multiple cases of normal lung, diffuse alveolar damage, COVID-19, influenza, and bacterial and fungal infections, as well as one case of pulmonary emboli, were included. The levels of CD61 staining were compared quantitatively in the autopsy cases, and patterns of staining were described. RESULTS Nearly all specimens exhibited an increase in CD61 staining relative to control lung tissue. The area of CD61 staining in COVID-19 infection was higher than influenza but still comparable to many other infectious diseases. Cases of aspiration pneumonia, Staphylococcus aureus infection, and blastomycosis exhibited the highest levels of CD61 staining. CONCLUSIONS Platelet deposition is a phenomenon common to many pulmonary insults. A spectrum of staining patterns was observed, suggestive of pathogen-specific mechanisms of platelet deposition. Further study into the mechanisms driving platelet deposition in pulmonary injuries and infections is warranted.
Collapse
Affiliation(s)
- Phillip D McMullen
- Department of Pathology, University of Chicago Medical Center, Chicago, IL
| | - Joseph H Cho
- Department of Pathology, University of Chicago Medical Center, Chicago, IL
| | - Jonathan L Miller
- Department of Pathology, University of Chicago Medical Center, Chicago, IL
| | - Aliya N Husain
- Department of Pathology, University of Chicago Medical Center, Chicago, IL
| | - Peter Pytel
- Department of Pathology, University of Chicago Medical Center, Chicago, IL
| | - Thomas Krausz
- Department of Pathology, University of Chicago Medical Center, Chicago, IL
| |
Collapse
|
17
|
Abstract
Obesity and obesity-related diseases like type 2 diabetes (T2D) are prominent global health issues; therefore, there is a need to better understand the mechanisms underlying these conditions. The onset of obesity is characterized by accumulation of proinflammatory cells, including Ly6chi monocytes (which differentiate into proinflammatory macrophages) and neutrophils, in metabolic tissues. This shift toward chronic, low-grade inflammation is an obese-state hallmark and highly linked to metabolic disorders and other obesity comorbidities. The mechanisms that induce and maintain increased inflammatory myelopoiesis are of great interest, with a recent focus on how obesity affects more primitive hematopoietic cells. The hematopoietic system is constantly replenished by proper regulation of hematopoietic stem and progenitor (HSPC) pools in the BM. While early research suggests that chronic obesity promotes expansion of myeloid-skewed HSPCs, the involvement of the hematopoietic stem cell (HSC) niche in regulating obesity-induced myelopoiesis remains undefined. In this review, we explore the role of the multicellular HSC niche in hematopoiesis and inflammation, and the potential contribution of this niche to the hematopoietic response to obesity. This review further aims to summarize the potential HSC niche involvement as a target of obesity-induced inflammation and a driver of obesity-induced myelopoiesis.
Collapse
|
18
|
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: 37] [Impact Index Per Article: 9.3] [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
|
19
|
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: 61] [Impact Index Per Article: 12.2] [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
|
20
|
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: 129] [Impact Index Per Article: 25.8] [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
|
21
|
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: 1.4] [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
|
22
|
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: 111] [Impact Index Per Article: 22.2] [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
|
23
|
Marín Oyarzún CP, Glembotsky AC, Goette NP, Lev PR, De Luca G, Baroni Pietto MC, Moiraghi B, Castro Ríos MA, Vicente A, Marta RF, Schattner M, Heller PG. Platelet Toll-Like Receptors Mediate Thromboinflammatory Responses in Patients With Essential Thrombocythemia. Front Immunol 2020; 11:705. [PMID: 32425934 PMCID: PMC7203216 DOI: 10.3389/fimmu.2020.00705] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/30/2020] [Indexed: 01/03/2023] Open
Abstract
Essential thrombocythemia (ET) is comprised among chronic myeloproliferative neoplasms (MPN) and is caused by driver mutations in JAK2, CALR, and MPL, which lead to megakaryocyte proliferation and prominent thrombocytosis. Thrombosis remains the main cause of morbidity in ET and is driven by the interplay between blood cells, the endothelium, the clotting cascade, and host-derived inflammatory mediators. Platelet activation plays a key role in the thrombotic predisposition, although the underlying mechanisms remain poorly defined. In addition to their role in hemostasis, platelets participate in innate immunity and inflammation owing to the expression of toll-like receptors (TLR), which recognize inflammatory signals, triggering platelet functional responses. Considering the impact of inflammation on ET procoagulant state, we assessed the contribution of TLR2 and TLR4 to platelet hemostatic and inflammatory properties in ET patients, by using Pam3CSK4 and lipopolysaccharide (LPS) as specific TLR2 and TLR4 ligands, respectively. TLR2 ligation induced increased surface translocation of α-granule-derived P-selectin and CD40L, which mediate platelet interaction with leukocytes and endothelial cells, respectively, and higher levels of dense granule-derived CD63 in patients, whereas PAC-1 binding was not increased and LPS had no effect on these platelet responses. Platelet-neutrophil aggregate formation was elevated in ET at baseline and after stimulation of both TLR2 and TLR4. In addition, ET patients displayed higher TLR2- and TLR4-triggered platelet secretion of the chemokine RANTES (CCL5), whereas von Willebrand factor release was not enhanced, revealing a differential releasate pattern for α-granule-stored inflammatory molecules. TLR-mediated hyperresponsiveness contrasted with impaired or preserved responses to classic platelet hemostatic agonists, such as TRAP-6 and thrombin. TLR2 and TLR4 expression on the platelet surface was normal, whereas phosphorylation of downstream effector ERK1/2 was higher in patients at baseline and after incubation with Pam3CSK4, which may partly explain the enhanced TLR2 response. In conclusion, exacerbated response to TLR stimulation may promote platelet activation in ET, boosting platelet/leukocyte/endothelial interactions and secretion of inflammatory mediators, overall reinforcing the thromboinflammatory state. These findings highlight the role of platelets as inflammatory sentinels in MPN prothrombotic scenario and provide additional evidence for the close intertwining between thrombosis and inflammation in this setting.
Collapse
Affiliation(s)
- Cecilia P Marín Oyarzún
- Institute of Medical Research A. Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Department of Hematology Research, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Ana C Glembotsky
- Institute of Medical Research A. Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Department of Hematology Research, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Nora P Goette
- Institute of Medical Research A. Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Paola R Lev
- Institute of Medical Research A. Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Department of Hematology Research, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Geraldine De Luca
- Institute of Medical Research A. Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Department of Hematology Research, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - María C Baroni Pietto
- Institute of Medical Research A. Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Department of Hematology Research, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Beatriz Moiraghi
- Department of Hematology, Hospital General de Agudos José María Ramos Mejía, Buenos Aires, Argentina
| | | | - Angeles Vicente
- Department of Hematology, Hospital Alemán, Buenos Aires, Argentina
| | - Rosana F Marta
- Institute of Medical Research A. Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Department of Hematology Research, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine (IMEX)- CONICET, National Academy of Medicine, Buenos Aires, Argentina
| | - Paula G Heller
- Institute of Medical Research A. Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina.,Department of Hematology Research, Institute of Medical Research (IDIM), National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
24
|
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: 6.4] [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
|
25
|
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: 1.3] [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
|
26
|
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: 2.3] [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
|
27
|
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: 13] [Impact Index Per Article: 2.2] [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
|
28
|
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: 160] [Impact Index Per Article: 26.7] [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
|
29
|
Abstract
Dysregulation of lymphocyte function, accumulation of autoantibodies and defective clearance of circulating immune complexes and apoptotic cells are hallmarks of systemic lupus erythematosus (SLE). Moreover, it is now evident that an intricate interplay between the adaptive and innate immune systems contributes to the pathogenesis of SLE, ultimately resulting in chronic inflammation and organ damage. Platelets circulate in the blood and are chiefly recognized for their role in the prevention of bleeding and promotion of haemostasis; however, accumulating evidence points to a role for platelets in both adaptive and innate immunity. Through a broad repertoire of receptors, platelets respond promptly to immune complexes, complement and damage-associated molecular patterns, and represent a major reservoir of immunomodulatory molecules in the circulation. Furthermore, evidence suggests that platelets are activated in patients with SLE, and that they could contribute to the circulatory autoantigenic load through the release of microparticles and mitochondrial antigens. Herein, we highlight how platelets contribute to the immune response and review evidence implicating platelets in the pathogenesis of SLE.
Collapse
|
30
|
PI3K/AKT and CD40L Signaling Regulate Platelet Activation and Endothelial Cell Damage in Sepsis. Inflammation 2019; 41:1815-1824. [PMID: 29956071 DOI: 10.1007/s10753-018-0824-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Platelets contribute to inflammation and their activation has been suggested as versatile effectors of sepsis. Activation of platelets promotes secretion of CD40L that induces sepsis and multiple organ dysfunction syndrome (MODS). However, the mechanisms regulate platelet-derived CD40L are not fully understood. Activation of PI3K/Akt pathway has been reported as a key component of sepsis, whereas the role of PI3K/Akt pathway in platelet-derived CD40L is unknown. In this study, we identified PI3K/Akt pathway as a key regulator of CD40L secretion by platelets. Significantly, inhibition of PI3K/Akt pathway by Ly294002 attenuated platelet activation and CD40L production. Moreover, PI3K/Akt pathway blocking suppresses vascular endothelial cells in vivo. Furthermore, the expression of biomarkers that represent the severity of sepsis, such as ICAM-1, VCAM-1, and E-selectin, was also suppressed by Ly294002. Altogether, our results confirm the pivotal role of PI3K/Akt pathway in sepsis and its inhibition might be a potential therapeutic target.
Collapse
|
31
|
The Role of Platelets in Antimicrobial Host Defense. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00029-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
|
32
|
Hamzeh-Cognasse H, Berthelot P, Tardy B, Pozzetto B, Bourlet T, Laradi S, Garraud O, Cognasse F. Platelet toll-like receptors are crucial sensors of infectious danger moieties. Platelets 2018; 29:533-540. [PMID: 29533683 DOI: 10.1080/09537104.2018.1445842] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/12/2018] [Accepted: 02/16/2018] [Indexed: 12/17/2022]
Abstract
In addition to their haemostatic role and function in the repair of damaged vascular epithelium, platelets play a defensive role in innate immunity, having the capacity to produce and secrete various anti-infectious factors, as well as cytokines, chemokines and related products, to interact with other immune cells to modulate immune responses to pathogens. Thus, it is now widely acknowledged that platelets participate in inflammatory processes and infection resolution, most notably by expressing and using receptors to bind infectious pathogen moieties and contributing to pathogen clearance. The ability of platelets to sense external danger signals relates to the expression of certain innate immunity receptors, such as toll-like receptors (TLRs), and the activation of efficient cell signalling machinery. TLR engagement triggers platelet response, which results in adapted degranulation according to: the type of TLR engaged, the nature of the ligand and the milieu; together, the TLR-mediated event and other signalling events may be followed by aggregation. Platelets thus use complex tools to mediate a whole range of functions upon sensing danger. By linking the inflammatory and haemostatic platelet response to infection, TLRs play a central role. The extent of the inflammatory response to pathogen clearance is still a debatable issue and is discussed in this short review.
Collapse
Affiliation(s)
| | - Philippe Berthelot
- a EA3064-GIMAP , University of Lyon-UJM , Saint-Etienne , France
- b Laboratory of Infectious Agents and Hygiene, University Hospital of Saint-Etienne , Saint Etienne , France
| | - Bernard Tardy
- c Clinical investigation Center-CIC 1408 , University Hospital of Saint-Etienne , Saint Etienne , France
- d Intensive Care Unit , University Hospital of Saint-Etienne , Saint Etienne , France
| | - Bruno Pozzetto
- a EA3064-GIMAP , University of Lyon-UJM , Saint-Etienne , France
- b Laboratory of Infectious Agents and Hygiene, University Hospital of Saint-Etienne , Saint Etienne , France
| | - Thomas Bourlet
- a EA3064-GIMAP , University of Lyon-UJM , Saint-Etienne , France
- b Laboratory of Infectious Agents and Hygiene, University Hospital of Saint-Etienne , Saint Etienne , France
| | - Sandrine Laradi
- a EA3064-GIMAP , University of Lyon-UJM , Saint-Etienne , France
- e EFS Auvergne-Rhône-Alpes , Saint-Etienne , France
| | - Olivier Garraud
- a EA3064-GIMAP , University of Lyon-UJM , Saint-Etienne , France
- f Institut National de la Transfusion Sanguine , Paris , France
| | - Fabrice Cognasse
- a EA3064-GIMAP , University of Lyon-UJM , Saint-Etienne , France
- e EFS Auvergne-Rhône-Alpes , Saint-Etienne , France
| |
Collapse
|
33
|
Deppermann C, Kubes P. Start a fire, kill the bug: The role of platelets in inflammation and infection. Innate Immun 2018; 24:335-348. [PMID: 30049243 PMCID: PMC6830908 DOI: 10.1177/1753425918789255] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/16/2018] [Accepted: 06/26/2018] [Indexed: 11/19/2022] Open
Abstract
Platelets are the main players in thrombosis and hemostasis; however they also play important roles during inflammation and infection. Through their surface receptors, platelets can directly interact with pathogens and immune cells. Platelets form complexes with neutrophils to modulate their capacities to produce reactive oxygen species or form neutrophil extracellular traps. Furthermore, they release microbicidal factors and cytokines that kill pathogens and influence the immune response, respectively. Platelets also maintain the vascular integrity during inflammation by a mechanism that is different from classical platelet activation. In this review we summarize the current knowledge about how platelets interact with the innate immune system during inflammation and infection and highlight recent advances in the field.
Collapse
Affiliation(s)
- Carsten Deppermann
- Calvin, Phoebe and Joan Snyder Institute for Chronic
Diseases, University of Calgary, Calgary, AB, Canada
| | - Paul Kubes
- Calvin, Phoebe and Joan Snyder Institute for Chronic
Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
34
|
Toll-like receptors in immunity and inflammatory diseases: Past, present, and future. Int Immunopharmacol 2018; 59:391-412. [PMID: 29730580 PMCID: PMC7106078 DOI: 10.1016/j.intimp.2018.03.002] [Citation(s) in RCA: 454] [Impact Index Per Article: 64.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 02/07/2023]
Abstract
The immune system is a very diverse system of the host that evolved during evolution to cope with various pathogens present in the vicinity of environmental surroundings inhabited by multicellular organisms ranging from achordates to chordates (including humans). For example, cells of immune system express various pattern recognition receptors (PRRs) that detect danger via recognizing specific pathogen-associated molecular patterns (PAMPs) and mount a specific immune response. Toll-like receptors (TLRs) are one of these PRRs expressed by various immune cells. However, they were first discovered in the Drosophila melanogaster (common fruit fly) as genes/proteins important in embryonic development and dorso-ventral body patterning/polarity. Till date, 13 different types of TLRs (TLR1-TLR13) have been discovered and described in mammals since the first discovery of TLR4 in humans in late 1997. This discovery of TLR4 in humans revolutionized the field of innate immunity and thus the immunology and host-pathogen interaction. Since then TLRs are found to be expressed on various immune cells and have been targeted for therapeutic drug development for various infectious and inflammatory diseases including cancer. Even, Single nucleotide polymorphisms (SNPs) among various TLR genes have been identified among the different human population and their association with susceptibility/resistance to certain infections and other inflammatory diseases. Thus, in the present review the current and future importance of TLRs in immunity, their pattern of expression among various immune cells along with TLR based therapeutic approach is reviewed. TLRs are first described PRRs that revolutionized the biology of host-pathogen interaction and immune response The discovery of different TLRs in humans proved milestone in the field of innate immunity and inflammation The pattern of expression of all the TLRs expressed by human immune cells An association of various TLR SNPs with different inflammatory diseases Currently available drugs or vaccines based on TLRs and their future in drug targeting along with the role in reproduction, and regeneration
Collapse
|
35
|
Claushuis TAM, Van Der Veen AIP, Horn J, Schultz MJ, Houtkooper RH, Van 't Veer C, Van Der Poll T. Platelet Toll-like receptor expression and activation induced by lipopolysaccharide and sepsis. Platelets 2018. [PMID: 29528268 DOI: 10.1080/09537104.2018.1445841] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Platelets and Toll-like receptor (TLR) signalling play a role in the immune response during sepsis. Although preclinical knowledge about the role of platelet TLR signalling is increasing, data during human sepsis are less abundant. Moreover, controversy remains about the effect of the TLR4 agonist lipopolysaccharide (LPS) on platelet activation. We therefore assessed platelet TLR expression during human and murine sepsis. Moreover, we investigated the effect of TLR4 signalling on platelet activation and TLR expression. Platelets from healthy controls stimulated with LPS did not show classical platelet activation (P-selectin, CD63 and phosphatidylserine expression), potentiation of subthreshold agonist stimulation nor platelet-leukocyte complex formation. LPS stimulation however did increase maximal mitochondrial respiration in a TLR4-dependent manner. Platelet stimulation with LPS did not alter TLR expression. Platelet stimulation with thrombin receptor activating peptide increased TLR5 and TLR9, but not TLR2 or TLR4 expression. Platelets from patients with sepsis and mice with experimental sepsis showed platelet activation, but unaltered TLR expression. These results indicate that sepsis-induced platelet activation is not associated with altered platelet TLR expression and, although platelets are responsive to LPS, stimulation of platelet TLR4 does not result in classical platelet activation.
Collapse
Affiliation(s)
- Theodora A M Claushuis
- a Center for Experimental and Molecular Medicine, Academic Medical Center , University of Amsterdam , Amsterdam , The Netherlands
| | - Annelou I P Van Der Veen
- b Department of Intensive Care, Academic Medical Center , University of Amsterdam , Amsterdam , The Netherlands
| | - Janneke Horn
- b Department of Intensive Care, Academic Medical Center , University of Amsterdam , Amsterdam , The Netherlands
| | - Marcus J Schultz
- b Department of Intensive Care, Academic Medical Center , University of Amsterdam , Amsterdam , The Netherlands
| | - Riekelt H Houtkooper
- c Laboratory Genetic Metabolic Diseases , Academic Medical Center , Amsterdam , The Netherlands.,d Amsterdam Institute for Gastroenterology and Metabolism (AG&M), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Cornelis Van 't Veer
- a Center for Experimental and Molecular Medicine, Academic Medical Center , University of Amsterdam , Amsterdam , The Netherlands
| | - Tom Van Der Poll
- a Center for Experimental and Molecular Medicine, Academic Medical Center , University of Amsterdam , Amsterdam , The Netherlands.,e Division of Infectious Diseases, Academic Medical Center , University of Amsterdam , Amsterdam , The Netherlands
| |
Collapse
|
36
|
Lopes Pires ME, Clarke SR, Marcondes S, Gibbins JM. Lipopolysaccharide potentiates platelet responses via toll-like receptor 4-stimulated Akt-Erk-PLA2 signalling. PLoS One 2017; 12:e0186981. [PMID: 29136006 PMCID: PMC5685579 DOI: 10.1371/journal.pone.0186981] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/11/2017] [Indexed: 02/06/2023] Open
Abstract
Lipopolysaccharide (LPS) from the cell envelope of Gram-negative bacteria is a principal cause of the symptoms of sepsis. LPS has been reported to modulate the function of platelets although the underlying mechanisms of LPS action in these cells remain unclear. Platelets express the Toll-like receptor 4 (TLR4) which serves as a receptor for LPS, although the potential role of TLR4 and associated cell signalling in controlling platelet responses to LPS has not been extensively explored. In this study, we therefore investigated the actions of LPS prepared from different strains of Escherichia coli on platelet function, the underlying signalling mechanisms, and the potential role of TLR4 in orchestrating these. We report that LPS increased the aggregation of washed platelets stimulated by thromboxane (U46619) or GPVI collagen receptor agonists, effects that were prevented by a TLR4 antagonist. Associated with this, LPS enhanced fibrinogen binding, P-selectin exposure and reactive oxygen species (ROS) release. Increase of ROS was found to be important for the actions of LPS on platelets, since these were inhibited in the presence of superoxide dismutase or catalase. The effects of LPS were associated with phosphorylation of Akt, ERK1/2 and PLA2 in stimulated platelets, and inhibitors of PI3-kinase, Akt and ERK1/2 reduced significantly LPS enhanced platelet function and associated ROS production. Furthermore, inhibition of platelet cyclooxygenase or the thromboxane receptor, revealed an important role for thromboxane A2. We therefore conclude that LPS increases human platelet activation through a TLR4-PI3K-Akt-ERK1/2-PLA2 -dependent pathway that is dependent on ROS and TXA2 formation.
Collapse
Affiliation(s)
- Maria E. Lopes Pires
- School of Biological Science, Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, Berkshire, United Kingdom
| | - Simon R. Clarke
- School of Biological Science, Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, Berkshire, United Kingdom
| | - Sisi Marcondes
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Jonathan M. Gibbins
- School of Biological Science, Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, Berkshire, United Kingdom
| |
Collapse
|
37
|
Toll-Like Receptor 4 Signalling and Its Impact on Platelet Function, Thrombosis, and Haemostasis. Mediators Inflamm 2017; 2017:9605894. [PMID: 29170605 PMCID: PMC5664350 DOI: 10.1155/2017/9605894] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/17/2017] [Accepted: 09/12/2017] [Indexed: 11/18/2022] Open
Abstract
Platelets are anucleated blood cells that participate in a wide range of physiological and pathological functions. Their major role is mediating haemostasis and thrombosis. In addition to these classic functions, platelets have emerged as important players in the innate immune system. In particular, they interact with leukocytes, secrete pro- and anti-inflammatory factors, and express a wide range of inflammatory receptors including Toll-like receptors (TLRs), for example, Toll-like receptor 4 (TLR4). TLR4, which is the most extensively studied TLR in nucleated cells, recognises lipopolysaccharides (LPS) that are compounds of the outer surface of Gram-negative bacteria. Unlike other TLRs, TLR4 is able to signal through both the MyD88-dependent and MyD88-independent signalling pathways. Notably, despite both pathways culminating in the activation of transcription factors, TLR4 has a prominent functional impact on platelet activity, haemostasis, and thrombosis. In this review, we summarise the current knowledge on TLR4 signalling in platelets, critically discuss its impact on platelet function, and highlight the open questions in this area.
Collapse
|
38
|
Brousseau-Nault M, Kizhakkedathu JN, Kim H. Chronic periodontitis is associated with platelet factor 4 (PF4) secretion: A pilot study. J Clin Periodontol 2017; 44:1101-1111. [DOI: 10.1111/jcpe.12771] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2017] [Indexed: 01/22/2023]
Affiliation(s)
- Mathieu Brousseau-Nault
- Faculty of Dentistry; University of British Columbia; Vancouver BC Canada
- Centre for Blood Research; University of British Columbia; Vancouver BC Canada
| | - Jayachandran N. Kizhakkedathu
- Centre for Blood Research; University of British Columbia; Vancouver BC Canada
- Department of Pathology and Laboratory Medicine; University of British Columbia; Vancouver BC Canada
- Department of Chemistry; University of British Columbia; Vancouver BC Canada
| | - Hugh Kim
- Faculty of Dentistry; University of British Columbia; Vancouver BC Canada
- Centre for Blood Research; University of British Columbia; Vancouver BC Canada
- Department of Biochemistry and Molecular Biology; University of British Columbia; Vancouver BC Canada
| |
Collapse
|
39
|
Megakaryocytes in Myeloproliferative Neoplasms Have Unique Somatic Mutations. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1512-1522. [PMID: 28502479 DOI: 10.1016/j.ajpath.2017.03.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/23/2017] [Indexed: 02/06/2023]
Abstract
Myeloproliferative neoplasms (MPNs) are a group of related clonal hemopoietic stem cell disorders associated with hyperproliferation of myeloid cells. They are driven by mutations in the hemopoietic stem cell, most notably JAK2V617F, CALR, and MPL. Clinically, they have the propensity to progress to myelofibrosis and transform to acute myeloid leukemia. Megakaryocytic hyperplasia with abnormal features are characteristic, and it is thought that these cells stimulate and drive fibrotic progression. The biological defects underpinning this remain to be explained. In this study we examined the megakaryocyte genome in 12 patients with MPNs to determine whether there are somatic variants and whether there is any association with marrow fibrosis. We performed targeted next-generation sequencing for 120 genes associated with myeloid neoplasms on megakaryocytes isolated from aspirated bone marrow. Ten of the 12 patients had genomic defects in megakaryocytes that were not present in nonmegakaryocytic hemopoietic marrow cells from the same patient. The greatest allelic burden was in patients with increased reticulin deposition. The megakaryocyte-unique mutations were predominantly in genes that regulate chromatin remodeling, chromosome alignment, and stability. These findings show that genomic abnormalities are present in megakaryocytes in MPNs and that these appear to be associated with progression to bone marrow fibrosis.
Collapse
|
40
|
Damien P, Cognasse F, Payrastre B, Spinelli SL, Blumberg N, Arthaud CA, Eyraud MA, Phipps RP, McNicol A, Pozzetto B, Garraud O, Hamzeh-Cognasse H. NF-κB Links TLR2 and PAR1 to Soluble Immunomodulator Factor Secretion in Human Platelets. Front Immunol 2017; 8:85. [PMID: 28220122 PMCID: PMC5292648 DOI: 10.3389/fimmu.2017.00085] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 01/18/2017] [Indexed: 12/13/2022] Open
Abstract
The primary toll-like receptor (TLR)-mediated immune cell response pathway common for all TLRs is MyD88-dependent activation of NF-κB, a seminal transcription factor for many chemokines and cytokines. Remarkably, anucleate platelets express the NF-κB machinery, whose role in platelets remains poorly understood. Here, we investigated the contribution of NF-κB in the release of cytokines and serotonin by human platelets, following selective stimulation of TLR2 and protease activated receptor 1 (PAR1), a classical and non-classical pattern-recognition receptor, respectively, able to participate to the innate immune system. We discovered that platelet PAR1 activation drives the process of NF-κB phosphorylation, in contrast to TLR2 activation, which induces a slower phosphorylation process. Conversely, platelet PAR1 and TLR2 activation induces similar ERK1/2, p38, and AKT phosphorylation. Moreover, we found that engagement of platelet TLR2 with its ligand, Pam3CSK4, significantly increases the release of sCD62P, RANTES, and sCD40L; this effect was attenuated by incubating platelets with a blocking anti-TLR2 antibody. This effect appeared selective since no modulation of serotonin secretion was observed following platelet TLR2 activation. Platelet release of sCD62P, RANTES, and sCD40L following TLR2 or PAR1 triggering was abolished in the presence of the NF-κB inhibitor Bay11-7082, while serotonin release following PAR1 activation was significantly decreased. These new findings support the concept that NF-κB is an important player in platelet immunoregulations and functions.
Collapse
Affiliation(s)
| | - Fabrice Cognasse
- GIMAP-EA3064, Université de Lyon, Saint-Étienne, France
- Etablissement Français du Sang Rhône-Alpes-Auvergne, Saint-Etienne, France
| | - Bernard Payrastre
- Inserm, U1048 and Université Toulouse 3, I2MC, CHU de Toulouse, Laboratoire d’Hématologie, Toulouse, France
| | - Sherry L. Spinelli
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Neil Blumberg
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | | | - Marie-Ange Eyraud
- Etablissement Français du Sang Rhône-Alpes-Auvergne, Saint-Etienne, France
| | - Richard P. Phipps
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | | | | | - Olivier Garraud
- GIMAP-EA3064, Université de Lyon, Saint-Étienne, France
- Institut National de Transfusion Sanguine (INTS), Paris, France
| | | |
Collapse
|
41
|
Abstract
The primary function of platelets is to patrol the vasculature and seal vessel breaches to limit blood loss. However, it is becoming increasingly clear that they also contribute to pathophysiological conditions like thrombosis, atherosclerosis, stroke and infection. Severe sepsis is a devastating disease that claims hundreds of thousands of lives every year in North America and is a major burden to the public health system. Platelet surface receptors like GPIb, αIIbβ3, TLR2 and TLR4 are involved in direct platelet-bacteria interactions. Plasma proteins like fibrinogen and vWF enable indirect interactions. Furthermore, platelet granules contain a plethora of proteins that modulate the immune response as well as microbicidal agents which can directly lyse bacteria. Bacterial toxins are potent platelet activators and can cause intravascular platelet aggregation. Platelets contribute to the antibacterial response of the host involving Kupffer cells, neutrophils and the complement system. In this review we summarize the current knowledge about platelet-bacteria interactions and highlight recent advances in the field.
Collapse
Affiliation(s)
- Carsten Deppermann
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Paul Kubes
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
42
|
Fuentes E, Rojas A, Palomo I. NF-κB signaling pathway as target for antiplatelet activity. Blood Rev 2016; 30:309-315. [PMID: 27075489 DOI: 10.1016/j.blre.2016.03.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 02/26/2016] [Accepted: 03/08/2016] [Indexed: 02/07/2023]
Abstract
In different nucleated cells, NF-κB has long been considered a prototypical proinflammatory signaling pathway with the expression of proinflammatory genes. Although platelets lack a nucleus, a number of functional transcription factors are involved in activated platelets, such as NF-κB. In platelet activation NF-κB regulation events include IKKβ phosphorylation, IκBα degradation, and p65 phosphorylation. Multiple pathways contribute to platelet activation and NF-κB is a common pathway in this activation. Therefore, in platelet activation the modulation of NF-κB pathway could be a potential new target in the treatment of inflammation-related vascular disease therapy (antiplatelet and antithrombotic activities).
Collapse
Affiliation(s)
- Eduardo Fuentes
- Laboratory of Hematology and Immunology, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001, Talca, Chile.
| | - Armando Rojas
- Biomedical Research Laboratories, Medicine Faculty, Catholic University of Maule, Talca, Chile
| | - Iván Palomo
- Laboratory of Hematology and Immunology, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001, Talca, Chile.
| |
Collapse
|
43
|
Abe H, Shiba M, Niibe Y, Tadokoro K, Satake M. Reduction of bacteria and human immunodeficiency virus Type 1 infectivity of platelet suspension in plasma using xenon flash-pulse light in a bench-scale trial. Transfusion 2016; 56:2256-66. [PMID: 27282889 DOI: 10.1111/trf.13685] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 03/09/2016] [Accepted: 05/02/2016] [Indexed: 01/19/2023]
Abstract
BACKGROUND Current pathogen reduction systems for platelet concentrates (PCs) require addition of chemical compounds and/or reduction of plasma content in PCs. We have investigated a new method using xenon (Xe) flash-pulse light without additional compounds or plasma replacement. STUDY DESIGN AND METHODS An aliquot of apheresis platelets (PLTs) in plasma inoculated with bacteria or human immunodeficiency virus Type 1 (HIV-1) was irradiated with Xe flash-pulse light (Xe flash phototreatment). Bacterial growth was monitored up to 6 days of storage, whereas HIV-1 infectivity was assayed just after treatment. Pairs of Xe flash-phototreated and untreated PCs were examined for PLT lesion during the storage period. RESULTS Under the current conditions, a low titer (1.8 colony-forming units [CFUs]/mL) of Staphylococcus aureus did not proliferate during the 6-day storage period, but grew in some cases at high-titer (24.0 CFUs/mL) inoculation. HIV-1 infectivity was reduced by 1.8 log. PLT recovery of the treated PCs was lower than untreated ones. An increase of mean PLT volume and glucose consumption, together with a decrease of hypotonic shock response and pH, were enhanced by the treatment. CD62P- and PAC-1-positive PLTs increased after the treatment, indicating the induction of PLT activation. Among biologic response modifiers, soluble CD40 ligand was significantly increased in the treated PCs on Day 6. CONCLUSIONS Xe flash phototreatment could prevent bacterial proliferation and reduce HIV-1 infectivity in 100% plasma PCs without any additional compounds, but enhanced PLT storage lesions. Further improvement is required to increase the potency of pathogen inactivation with reducing PLT damage.
Collapse
Affiliation(s)
- Hideki Abe
- Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan.
| | - Masayuki Shiba
- Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| | | | - Kenji Tadokoro
- Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| | - Masahiro Satake
- Central Blood Institute, Blood Service Headquarters, Japanese Red Cross Society, Tokyo, Japan
| |
Collapse
|
44
|
Nagasawa T, Somamoto T, Nakao M. Carp thrombocyte phagocytosis requires activation factors secreted from other leukocytes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 52:107-111. [PMID: 25978929 DOI: 10.1016/j.dci.2015.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/07/2015] [Accepted: 05/08/2015] [Indexed: 06/04/2023]
Abstract
Thrombocytes are nucleated blood cells in non-mammalian vertebrates, which were recently focused on not only as hemostatic cells but also as immune cells with potent phagocytic activities. We have analyzed the phagocytic activation mechanisms in common carp (Cyprinus carpio) thrombocytes. MACS-sorted mAb(+) thrombocytes showed no phagocytic activity even in the presence of several stimulants. However, remixing these thrombocytes with other anti-thrombocyte mAb(-) leukocyte populations restored their phagocytic activities, indicating that carp thrombocyte phagocytosis requires an appropriate exogenous stimulation. Culture supernatant from anti-thrombocyte mAb(-) leukocytes harvested after PMA or LPS stimulation, but not culture supernatant from unstimulated leukocytes, could activate thrombocyte phagocytosis. This proposed mechanism of thrombocyte phagocytosis activation involving soluble factors produced by activated leukocytes suggests that thrombocyte activation is restricted to areas proximal to injured tissues, ensuring suppression of excessive thrombocyte activation and a balance between inflammation and tissue repair.
Collapse
Affiliation(s)
- Takahiro Nagasawa
- Laboratory of Marine Biochemistry, Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 812-8581, Japan
| | - Tomonori Somamoto
- Laboratory of Marine Biochemistry, Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 812-8581, Japan.
| | - Miki Nakao
- Laboratory of Marine Biochemistry, Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 812-8581, Japan
| |
Collapse
|
45
|
Reviakine I. New horizons in platelet research: Understanding and harnessing platelet functional diversity. Clin Hemorheol Microcirc 2015; 60:133-52. [DOI: 10.3233/ch-151942] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
46
|
Nguyen KA, Hamzeh-Cognasse H, Laradi S, Pozzetto B, Garraud O, Cognasse F. Specific activation, signalling and secretion profiles of human platelets following PAR-1 and PAR-4 stimulation. Platelets 2015; 26:795-798. [PMID: 26083907 DOI: 10.3109/09537104.2015.1050585] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Blood platelets play a central haemostatic function; however, they also play a role in inflammation and are capable of secreting various cytokines, chemokines and related products. The purpose of this study was to identify subtle variations in platelet physiology using proteomics. We compared the levels of membrane proteins (n = 3), α and δ granule proteins (n = 18), and signalling proteins (n = 30) from unstimulated platelets with those of protease-activated receptor (PAR)-1- and PAR-4-stimulated platelets (n = 10). The vast majority of these proteins responded similarly to PAR-1 or PAR-4 engagement. However, differences were observed within membrane CD40L expressed, and α granule GRO-α and MDC secreted proteins.
Collapse
Affiliation(s)
| | | | - Sandrine Laradi
- a Université de Lyon , Saint Etienne , France
- b Etablissement Français du Sang - Auvergne-Loire , Saint-Etienne , France , and
| | | | - Olivier Garraud
- a Université de Lyon , Saint Etienne , France
- c INTS - Institut National de la Transfusion Sanguine , Paris , France
| | - Fabrice Cognasse
- a Université de Lyon , Saint Etienne , France
- b Etablissement Français du Sang - Auvergne-Loire , Saint-Etienne , France , and
| |
Collapse
|
47
|
Loh YS, Dean MM, Johnson L, Marks DC. Treatment of platelets with riboflavin and ultraviolet light mediates complement activation and suppresses monocyte interleukin-12 production in whole blood. Vox Sang 2015; 109:327-35. [DOI: 10.1111/vox.12283] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 03/03/2015] [Accepted: 03/17/2015] [Indexed: 01/07/2023]
Affiliation(s)
- Y. S. Loh
- Research and Development; Australian Red Cross Blood Service; Sydney NSW Australia
| | - M. M. Dean
- Research and Development; Australian Red Cross Blood Service; Brisbane QLD Australia
| | - L. Johnson
- Research and Development; Australian Red Cross Blood Service; Sydney NSW Australia
| | - D. C. Marks
- Research and Development; Australian Red Cross Blood Service; Sydney NSW Australia
| |
Collapse
|
48
|
Abstract
Bacteria that enter the bloodstream will encounter components of the cellular and soluble immune response. Platelets contribute to this response and have emerged as an important target for bacterial pathogens. Bacteria produce diverse extracellular proteins and toxins that have been reported to modulate platelet function. These interactions can result in complete or incomplete platelet activation or inhibition of platelet activation, depending on the bacteria and bacterial product. The nature of the platelet response may be highly relevant to disease pathogenesis.
Collapse
Affiliation(s)
- Oonagh Shannon
- Division of Infection Medicine, Department of Clinical Sciences, Lund University , Lund , Sweden
| |
Collapse
|
49
|
Abstract
Multiple studies have now shown that various species of bacteria can stimulate platelets; many in a strain and donor-dependent manner. The signalling pathways underlying this platelet activation has been the subject of scrutiny for the last decade. The best-delineated pathway is that in response to Streptococcal species, such as Streptococcus sanguinis (S. sanguinis), Streptococcus gordonii (S. gordonii) and Streptococcus oralis (S. oralis), where a pathway is initiated by the engagement of the low affinity IgG receptor, FcγRIIA. This leads to and involves the tyrosine kinase Syk, the adaptor protein Linker of Activated T Cells (LAT) and subsequently both phospholipase Cγ2 (PLCγ2) and phosphatidylinositol-3-kinase (PI-3-K). Finally, this leads to the expression of the αIIbβ3 integrin, the synthesis and release of thromboxane A2 (T × A2) and the exocytosis of PF4, each of which plays a crucial role in secondary signalling and full platelet activation. Roles for other signalling pathways in Streptococcal-induced platelet activation are less clear, although an ADP-mediated inhibition of adenylyl cyclase, a glycoprotein Ib/IX/V-mediated pathway and perhaps a complement-induced pathway have each been proposed. Platelet activation by Porphyromonas gingivalis (P. gingivalis) at least partially shares the FcγRIIA/Syk/PLCγ2/PI-3-K mechanism utilised by Streptococcal species. However, it has also been suggested that P. gingivalis activates platelets by two additional methods; stimulation of the protease-activated receptors leading to activation of phospholipase Cβ (PLCβ), and the engagement of Toll-like receptors 2 and 4 by released lipopolysaccharide leading to an ill-defined pathway which may involve PI-3-K. Consequently, it appears that bacteria can stimulate platelets by eliciting multiple signalling pathways some of which are common, and some unique, to individual species.
Collapse
|
50
|
Nording HM, Seizer P, Langer HF. Platelets in inflammation and atherogenesis. Front Immunol 2015; 6:98. [PMID: 25798138 PMCID: PMC4351644 DOI: 10.3389/fimmu.2015.00098] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 02/19/2015] [Indexed: 12/12/2022] Open
Abstract
Platelets contribute to processes beyond thrombus formation and may play a so far underestimated role as an immune cell in various circumstances. This review outlines immune functions of platelets in host defense, but also how they may contribute to mechanisms of infectious diseases. A particular emphasis is placed on the interaction of platelets with other immune cells. Furthermore, this article outlines the features of atherosclerosis as an inflammatory vascular disease highlighting the role of platelet crosstalk with cellular and soluble factors involved in atheroprogression. Understanding, how platelets influence these processes of vascular remodeling will shed light on their role for tissue homeostasis beyond intravascular thrombosis. Finally, translational implications of platelet-mediated inflammation in atherosclerosis are discussed.
Collapse
Affiliation(s)
- Henry M. Nording
- University Clinic for Cardiology and Cardiovascular Medicine, Eberhard Karls-University Tübingen, Tübingen, Germany
- Section for Cardioimmunology, Eberhard Karls-University Tübingen, Tübingen, Germany
| | - Peter Seizer
- University Clinic for Cardiology and Cardiovascular Medicine, Eberhard Karls-University Tübingen, Tübingen, Germany
| | - Harald F. Langer
- University Clinic for Cardiology and Cardiovascular Medicine, Eberhard Karls-University Tübingen, Tübingen, Germany
- Section for Cardioimmunology, Eberhard Karls-University Tübingen, Tübingen, Germany
| |
Collapse
|