1
|
Zhou Y, Dong J, Wang M, Liu Y. New insights of platelet endocytosis and its implication for platelet function. Front Cardiovasc Med 2024; 10:1308170. [PMID: 38264257 PMCID: PMC10803655 DOI: 10.3389/fcvm.2023.1308170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/21/2023] [Indexed: 01/25/2024] Open
Abstract
Endocytosis constitutes a cellular process in which cells selectively encapsulate surface substances into endocytic vesicles, also known as endosomes, thereby modulating their interaction with the environment. Platelets, as pivotal hematologic elements, play a crucial role not only in regulating coagulation and thrombus formation but also in facilitating tumor invasion and metastasis. Functioning as critical components in the circulatory system, platelets can internalize various endosomal compartments, such as surface receptors, extracellular proteins, small molecules, and pathogens, from the extracellular environment through diverse endocytic pathways, including pinocytosis, phagocytosis, and receptor-mediated endocytosis. We summarize recent advancements in platelet endocytosis, encompassing the catalog of cargoes, regulatory mechanisms, and internal trafficking routes. Furthermore, we describe the influence of endocytosis on platelet regulatory functions and related physiological and pathological processes, aiming to offer foundational insights for future research into platelet endocytosis.
Collapse
Affiliation(s)
- Yangfan Zhou
- Department of Cardiology, Cardiovascular Center, Henan Key Laboratory of Hereditary Cardiovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jianzeng Dong
- Department of Cardiology, Cardiovascular Center, Henan Key Laboratory of Hereditary Cardiovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- National Clinical Research Centre for Cardiovascular Diseases, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Mengyu Wang
- Department of Cardiology, Cardiovascular Center, Henan Key Laboratory of Hereditary Cardiovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yangyang Liu
- Department of Cardiology, Cardiovascular Center, Henan Key Laboratory of Hereditary Cardiovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
2
|
Awamura T, Nakasone ES, Gangcuangco LM, Subia NT, Bali AJ, Chow DC, Shikuma CM, Park J. Platelet and HIV Interactions and Their Contribution to Non-AIDS Comorbidities. Biomolecules 2023; 13:1608. [PMID: 38002289 PMCID: PMC10669125 DOI: 10.3390/biom13111608] [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: 10/05/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Platelets are anucleate cytoplasmic cell fragments that circulate in the blood, where they are involved in regulating hemostasis. Beyond their normal physiologic role, platelets have emerged as versatile effectors of immune response. During an infection, cell surface receptors enable platelets to recognize viruses, resulting in their activation. Activated platelets release biologically active molecules that further trigger host immune responses to protect the body against infection. Their impact on the immune response is also associated with the recruitment of circulating leukocytes to the site of infection. They can also aggregate with leukocytes, including lymphocytes, monocytes, and neutrophils, to immobilize pathogens and prevent viral dissemination. Despite their host protective role, platelets have also been shown to be associated with various pathophysiological processes. In this review, we will summarize platelet and HIV interactions during infection. We will also highlight and discuss platelet and platelet-derived mediators, how they interact with immune cells, and the multifaceted responsibilities of platelets in HIV infection. Furthermore, we will give an overview of non-AIDS comorbidities linked to platelet dysfunction and the impact of antiretroviral therapy on platelet function.
Collapse
Affiliation(s)
- Thomas Awamura
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
| | - Elizabeth S. Nakasone
- University of Hawai‘i Cancer Center, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA;
- Department of Medicine, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA;
| | - Louie Mar Gangcuangco
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| | - Natalie T. Subia
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
| | - Aeron-Justin Bali
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
| | - Dominic C. Chow
- Department of Medicine, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA;
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| | - Cecilia M. Shikuma
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| | - Juwon Park
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| |
Collapse
|
3
|
Ferdous F, Scott T. The Immunological Capacity of Thrombocytes. Int J Mol Sci 2023; 24:12950. [PMID: 37629130 PMCID: PMC10454457 DOI: 10.3390/ijms241612950] [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: 06/22/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Thrombocytes are numerous in the blood of aves (birds) and ichthyoids (fish). The origin of this cell type is a common hematopoietic stem cell giving rise to a cell that is active in blood coagulation, inflammatory functions, and the immune response in general. It has been well documented that thrombocytes can phagocytize small particles and bacteria. While phagocytosis with an associated oxidative burst has been reported for chicken thrombocytes, some questions remain as to the degradation capacity of phagosomes in ichthyoids. As innate cells, thrombocytes can be stimulated by bacterial, viral, and fungal pathogens to express altered gene expression. Furthermore, there have been observations that led researchers to state that platelets/thrombocytes are capable of serving as "professional antigen presenting cells" expressing CD40, CD80/86, MHC I, and MHC II. This indeed may be the case or, more likely at this time, provide supporting evidence that these cells aid and assist in the role of professional antigen-presenting cells to initiate adaptive immune responses.
Collapse
Affiliation(s)
- Farzana Ferdous
- Department of Biological Sciences, University of North Carolina at Charlotte, 9201 University City Blvd., Charlotte, NC 28223, USA
| | - Thomas Scott
- Department of Animal & Veterinary Sciences, Clemson University, 129 Poole Agricultural Center, Clemson, SC 29634, USA;
| |
Collapse
|
4
|
Tovo PA, Galliano I, Parodi E, Calvi C, Gambarino S, Licciardi F, Dini M, Montanari P, Branca M, Ramenghi U, Bergallo M. Children with Chronic Immune Thrombocytopenia Exhibit High Expression of Human Endogenous Retroviruses TRIM28 and SETDB1. Genes (Basel) 2023; 14:1569. [PMID: 37628621 PMCID: PMC10454145 DOI: 10.3390/genes14081569] [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: 07/11/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Chronic immune thrombocytopenia (CITP) is an autoimmune disease whose underlying biologic mechanisms remain elusive. Human endogenous retroviruses (HERVs) derive from ancestral infections and constitute about 8% of our genome. A wealth of clinical and experimental studies highlights their pivotal pathogenetic role in autoimmune diseases. Epigenetic mechanisms, such as those modulated by TRIM28 and SETDB1, are involved in HERV activation and regulation of immune response. We assessed, through a polymerase chain reaction real-time Taqman amplification assay, the transcription levels of pol genes of HERV-H, HERV-K, and HERV-W; env genes of Syncytin (SYN)1, SYN2, and HERV-W; as well as TRIM28 and SETDB1 in whole blood from 34 children with CITP and age-matched healthy controls (HC). The transcriptional levels of all HERV sequences, with the exception of HERV-W-env, were significantly enhanced in children with CITP as compared to HC. Patients on eltrombopag treatment exhibited lower expression of SYN1, SYN2, and HERV-W-env as compared to untreated patients. The mRNA concentrations of TRIM28 and SETDB1 were significantly higher and were positively correlated with those of HERVs in CITP patients. The over-expressions of HERVs and TRIM28/SETDB1 and their positive correlations in patients with CITP are suggestive clues of their contribution to the pathogenesis of the disease and support innovative interventions to inhibit HERV and TRIM28/SETDB1 expressions in patients unresponsive to standard therapies.
Collapse
Affiliation(s)
- Pier-Angelo Tovo
- Department of Public Health and Pediatric Sciences, University of Turin, Piazza Polonia 94, 10126 Turin, Italy; (P.-A.T.); (U.R.)
| | - Ilaria Galliano
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, Regina Margherita Children’s Hospitalno, Piazza Polonia 94, 10126 Turin, Italy; (I.G.); (C.C.); (S.G.); (M.D.); (P.M.)
| | - Emilia Parodi
- Pediatric and Neonatology Unit, Ordine Mauriziano Hospital, Largo Filippo Turati 62, 10128 Turin, Italy;
| | - Cristina Calvi
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, Regina Margherita Children’s Hospitalno, Piazza Polonia 94, 10126 Turin, Italy; (I.G.); (C.C.); (S.G.); (M.D.); (P.M.)
| | - Stefano Gambarino
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, Regina Margherita Children’s Hospitalno, Piazza Polonia 94, 10126 Turin, Italy; (I.G.); (C.C.); (S.G.); (M.D.); (P.M.)
| | - Francesco Licciardi
- Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy;
| | - Maddalena Dini
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, Regina Margherita Children’s Hospitalno, Piazza Polonia 94, 10126 Turin, Italy; (I.G.); (C.C.); (S.G.); (M.D.); (P.M.)
| | - Paola Montanari
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, Regina Margherita Children’s Hospitalno, Piazza Polonia 94, 10126 Turin, Italy; (I.G.); (C.C.); (S.G.); (M.D.); (P.M.)
| | - Margherita Branca
- Postgraduate School of Pediatrics, University of Turin, Piazza Polonia 94, 10126 Turin, Italy;
| | - Ugo Ramenghi
- Department of Public Health and Pediatric Sciences, University of Turin, Piazza Polonia 94, 10126 Turin, Italy; (P.-A.T.); (U.R.)
- Regina Margherita Children’s Hospital, Piazza Polonia 94, 10126 Turin, Italy;
- Postgraduate School of Pediatrics, University of Turin, Piazza Polonia 94, 10126 Turin, Italy;
| | - Massimiliano Bergallo
- Pediatric Laboratory, Department of Public Health and Pediatric Sciences, University of Turin, Regina Margherita Children’s Hospitalno, Piazza Polonia 94, 10126 Turin, Italy; (I.G.); (C.C.); (S.G.); (M.D.); (P.M.)
| |
Collapse
|
5
|
Zhang Z, Zhou XH, Cheng ZP, Hu Y. [Research on immunological function of platelet receptor FcγRⅡA]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2023; 44:609-614. [PMID: 37749049 PMCID: PMC10509618 DOI: 10.3760/cma.j.issn.0253-2727.2023.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Indexed: 09/27/2023]
Affiliation(s)
- Z Zhang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - X H Zhou
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Z P Cheng
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Y Hu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
6
|
Ragnoli B, Da Re B, Galantino A, Kette S, Salotti A, Malerba M. Interrelationship between COVID-19 and Coagulopathy: Pathophysiological and Clinical Evidence. Int J Mol Sci 2023; 24:ijms24108945. [PMID: 37240292 DOI: 10.3390/ijms24108945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/14/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Since the first description of COVID-19 infection, among clinical manifestations of the disease, including fever, dyspnea, cough, and fatigue, it was observed a high incidence of thromboembolic events potentially evolving towards acute respiratory distress syndrome (ARDS) and COVID-19-associated-coagulopathy (CAC). The hypercoagulation state is based on an interaction between thrombosis and inflammation. The so-called CAC represents a key aspect in the genesis of organ damage from SARS-CoV-2. The prothrombotic status of COVID-19 can be explained by the increase in coagulation levels of D-dimer, lymphocytes, fibrinogen, interleukin 6 (IL-6), and prothrombin time. Several mechanisms have been hypothesized to explain this hypercoagulable process such as inflammatory cytokine storm, platelet activation, endothelial dysfunction, and stasis for a long time. The purpose of this narrative review is to provide an overview of the current knowledge on the pathogenic mechanisms of coagulopathy that may characterize COVID-19 infection and inform on new areas of research. New vascular therapeutic strategies are also reviewed.
Collapse
Affiliation(s)
| | - Beatrice Da Re
- Respiratory Unit, Sant'Andrea Hospital, 13100 Vercelli, Italy
| | | | - Stefano Kette
- Respiratory Unit, Sant'Andrea Hospital, 13100 Vercelli, Italy
| | - Andrea Salotti
- Respiratory Unit, Sant'Andrea Hospital, 13100 Vercelli, Italy
| | - Mario Malerba
- Respiratory Unit, Sant'Andrea Hospital, 13100 Vercelli, Italy
- Department of Traslational Medicine, University of Eastern Piedmont (UPO), 28100 Novara, Italy
| |
Collapse
|
7
|
Garraud O, Hamzeh-Cognasse H, Chalayer E, Duchez AC, Tardy B, Oriol P, Haddad A, Guyotat D, Cognasse F. Platelet transfusion in adults: An update. Transfus Clin Biol 2023; 30:147-165. [PMID: 36031180 DOI: 10.1016/j.tracli.2022.08.147] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Many patients worldwide receive platelet components (PCs) through the transfusion of diverse types of blood components. PC transfusions are essential for the treatment of central thrombocytopenia of diverse causes, and such treatment is beneficial in patients at risk of severe bleeding. PC transfusions account for almost 10% of all the blood components supplied by blood services, but they are associated with about 3.25 times as many severe reactions (attributable to transfusion) than red blood cell transfusions after stringent in-process leukoreduction to less than 106 residual cells per blood component. PCs are not homogeneous, due to the considerable differences between donors. Furthermore, the modes of PC collection and preparation, the safety precautions taken to limit either the most common (allergic-type reactions and febrile non-hemolytic reactions) or the most severe (bacterial contamination, pulmonary lesions) adverse reactions, and storage and conservation methods can all result in so-called PC "storage lesions". Some storage lesions affect PC quality, with implications for patient outcome. Good transfusion practices should result in higher levels of platelet recovery and efficacy, and lower complication rates. These practices include a matching of tissue ABH antigens whenever possible, and of platelet HLA (and, to a lesser extent, HPA) antigens in immunization situations. This review provides an overview of all the available information relating to platelet transfusion, from donor and donation to bedside transfusion, and considers the impact of the measures applied to increase transfusion efficacy while improving safety and preventing transfusion inefficacy and refractoriness. It also considers alternatives to platelet component (PC) transfusion.
Collapse
Affiliation(s)
- O Garraud
- SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Étienne, France.
| | | | - E Chalayer
- SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Étienne, France; Saint-Etienne University Hospital, Department of Hematology and Cellular Therapy, Saint-Étienne, France
| | - A C Duchez
- SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Étienne, France; Établissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
| | - B Tardy
- SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Étienne, France; CHU de Saint-Etienne, INSERM and CIC EC 1408, Clinical Epidemiology, Saint-Étienne, France
| | - P Oriol
- CHU de Saint-Etienne, INSERM and CIC EC 1408, Clinical Epidemiology, Saint-Étienne, France
| | - A Haddad
- SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Étienne, France; Sacré-Cœur Hospital, Beirut, Lebanon; Lebanese American University, Beirut, Lebanon
| | - D Guyotat
- Saint-Etienne University Hospital, Department of Hematology and Cellular Therapy, Saint-Étienne, France
| | - F Cognasse
- SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Étienne, France; Établissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
| |
Collapse
|
8
|
Sharma S, Tyagi T, Antoniak S. Platelet in thrombo-inflammation: Unraveling new therapeutic targets. Front Immunol 2022; 13:1039843. [PMID: 36451834 PMCID: PMC9702553 DOI: 10.3389/fimmu.2022.1039843] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
In the broad range of human diseases, thrombo-inflammation appears as a clinical manifestation. Clinically, it is well characterized in context of superficial thrombophlebitis that is recognized as thrombosis and inflammation of superficial veins. However, it is more hazardous when developed in the microvasculature of injured/inflamed/infected tissues and organs. Several diseases like sepsis and ischemia-reperfusion can cause formation of microvascular thrombosis subsequently leading to thrombo-inflammation. Thrombo-inflammation can also occur in cases of antiphospholipid syndrome, preeclampsia, sickle cell disease, bacterial and viral infection. One of the major contributors to thrombo-inflammation is the loss of normal anti-thrombotic and anti-inflammatory potential of the endothelial cells of vasculature. This manifest itself in the form of dysregulation of the coagulation pathway and complement system, pathologic platelet activation, and increased recruitment of leukocyte within the microvasculature. The role of platelets in hemostasis and formation of thrombi under pathologic and non-pathologic conditions is well established. Platelets are anucleate cells known for their essential role in primary hemostasis and the coagulation pathway. In recent years, studies provide strong evidence for the critical involvement of platelets in inflammatory processes like acute ischemic stroke, and viral infections like Coronavirus disease 2019 (COVID-19). This has encouraged the researchers to investigate the contribution of platelets in the pathology of various thrombo-inflammatory diseases. The inhibition of platelet surface receptors or their intracellular signaling which mediate initial platelet activation and adhesion might prove to be suitable targets in thrombo-inflammatory disorders. Thus, the present review summarizes the concept and mechanism of platelet signaling and briefly discuss their role in sterile and non-sterile thrombo-inflammation, with the emphasis on role of platelets in COVID-19 induced thrombo-inflammation. The aim of this review is to summarize the recent developments in deciphering the role of the platelets in thrombo-inflammation and discuss their potential as pharmaceutical targets.
Collapse
Affiliation(s)
- Swati Sharma
- UNC Blood Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Tarun Tyagi
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| | - Silvio Antoniak
- UNC Blood Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
9
|
Losada PX, DeLaura I, Narváez CF. Dengue Virus and Platelets: From the Biology to the Clinic. Viral Immunol 2022; 35:349-358. [PMID: 35483090 DOI: 10.1089/vim.2021.0135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Dengue is one of the most important vector-borne viral illnesses found in tropical and subtropical regions. Colombia has one of the highest rates of dengue cases in the Americas. Severe dengue virus (DENV) infection presents with capillary leakage, hemorrhage, and organ compromise, eventually leading to death. Over the years, there have been many efforts to develop a vaccine that guarantees protective immunity, but they have been partially successful, as such immunity would need to guarantee protection against four distinct viral serotypes. Absolute platelet count is a laboratory parameter used to monitor the clinical progression of DENV, as infection is often accompanied by thrombocytopenia. Although this finding is well described with respect to the natural history of the disease, there are various hypotheses as to the cause of this rapid decrease, and several in vivo and ex vivo models have been used to explain the effect of DENV infection on platelets and their precursors. DENV infects and activates platelets, facilitating their elimination through recognition by phagocytic cells and peripheral margination. However, infection also affects the precursors in the bone marrow by modulating megakaryopoiesis. The objective of this article is to explore various proposed mechanisms of DENV-induced thrombocytopenia to better understand the pathophysiology and clinical presentations of this highly relevant viral infection.
Collapse
Affiliation(s)
- Paula X Losada
- División de Inmunología, Programa de Medicina, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - Isabel DeLaura
- Duke University School of Medicine, Durham, North Carolina, USA
| | - Carlos F Narváez
- División de Inmunología, Programa de Medicina, Facultad de Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
| |
Collapse
|
10
|
Thiele T, Schwarz SL, Handtke S. Platelet size as a mirror for the immune response after SARS-CoV-2 vaccination. J Thromb Haemost 2022; 20:818-820. [PMID: 35156282 PMCID: PMC9115134 DOI: 10.1111/jth.15659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 01/26/2022] [Accepted: 01/26/2022] [Indexed: 11/18/2022]
Affiliation(s)
- Thomas Thiele
- Institut für Immunologie und Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Silas L Schwarz
- Institut für Immunologie und Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Stefan Handtke
- Institut für Immunologie und Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
11
|
Cognasse F, Duchez AC, Audoux E, Ebermeyer T, Arthaud CA, Prier A, Eyraud MA, Mismetti P, Garraud O, Bertoletti L, Hamzeh-Cognasse H. Platelets as Key Factors in Inflammation: Focus on CD40L/CD40. Front Immunol 2022; 13:825892. [PMID: 35185916 PMCID: PMC8850464 DOI: 10.3389/fimmu.2022.825892] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/14/2022] [Indexed: 12/16/2022] Open
Abstract
Platelets are anucleate cytoplasmic fragments derived from the fragmentation of medullary megakaryocytes. Activated platelets adhere to the damaged endothelium by means of glycoproteins on their surface, forming the platelet plug. Activated platelets can also secrete the contents of their granules, notably the growth factors contained in the α-granules, which are involved in platelet aggregation and maintain endothelial activation, but also contribute to vascular repair and angiogenesis. Platelets also have a major inflammatory and immune function in antibacterial defence, essentially through their Toll-like Receptors (TLRs) and Sialic acid-binding immunoglobulin-type lectin (SIGLEC). Platelet activation also contributes to the extensive release of anti- or pro-inflammatory mediators such as IL-1β, RANTES (Regulated on Activation, Normal T Expressed and Secreted) or CD154, also known as the CD40-ligand. Platelets are involved in the direct activation of immune cells, polynuclear neutrophils (PNNs) and dendritic cells via the CD40L/CD40 complex. As a general rule, all of the studies presented in this review show that platelets are capable of covering most of the stages of inflammation, primarily through the CD40L/CD40 interaction, thus confirming their own role in this pathophysiological condition.
Collapse
Affiliation(s)
- Fabrice Cognasse
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France.,SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Anne Claire Duchez
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France.,SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Estelle Audoux
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France.,SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Theo Ebermeyer
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France.,SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Charles Antoine Arthaud
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France.,SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Amelie Prier
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France.,SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Marie Ange Eyraud
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France.,SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Patrick Mismetti
- SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France.,Vascular and Therapeutic Medicine Department, Saint-Etienne University Hospital Center, Saint-Etienne, France
| | - Olivier Garraud
- SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France
| | - Laurent Bertoletti
- SAINBIOSE, INSERM, U1059, University of Lyon, Saint-Etienne, France.,Vascular and Therapeutic Medicine Department, Saint-Etienne University Hospital Center, Saint-Etienne, France
| | | |
Collapse
|
12
|
Georgakopoulou VE, Lembessis P, Skarlis C, Gkoufa A, Sipsas NV, Mavragani CP. Hematological Abnormalities in COVID-19 Disease: Association With Type I Interferon Pathway Activation and Disease Outcomes. Front Med (Lausanne) 2022; 9:850472. [PMID: 35372456 PMCID: PMC8968418 DOI: 10.3389/fmed.2022.850472] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/21/2022] [Indexed: 12/29/2022] Open
Abstract
Increased expression of interferon (IFN)-stimulated genes (ISGs) in peripheral blood, has been previously reported in viral infections, as well as in autoimmune disorders, in association with reduced leukocyte and platelet counts. Though cytopenias are common in patients with COVID-19 disease and predict severe outcomes, the underlying mechanisms have not been fully elucidated. In the current study, we aimed to determine the prevalence of hematological abnormalities in the setting of active COVID-19 infection and to explore whether they associate with disease outcomes and activation of type I IFN pathway. One-hundred-twenty-three consecutive SARS-CoV2 infected patients were included in the study. Clinical and laboratory parameters were recorded for all study participants. In 114 patients, total RNA was extracted from whole peripheral blood and subjected to real time PCR. The relative expression of three interferon stimulated genes (ISGs; IFIT1, MX-1, and IFI44) was determined and a type I IFN score reflecting peripheral type I IFN activity was calculated. The rates of anemia, leukopenia, and thrombocytopenia were 28.5, 14.6, and 24.4%, respectively. Among leukocytopenias, eosinopenia, and lymphopenia were the most prominent abnormalities being found in 56.9 and 43.1%, respectively. Of interest, patients with either eosinopenia and/or thrombocytopenia but no other hematological abnormalities displayed significantly increased peripheral type I IFN scores compared to their counterparts with normal/high eosinophil and platelet counts. While eosinopenia along with lymphopenia were found to be associated with increased risk for intubation and severe/critical disease, such an association was not detected between other hematological abnormalities or increased type I IFN scores. In conclusion, hematological abnormalities are commonly detected among patients with COVID-19 infection in association with severe disease outcomes and activation of the type I IFN pathway.
Collapse
Affiliation(s)
- Vasiliki E. Georgakopoulou
- Department of Infectious Diseases and COVID-19 Unit, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Lembessis
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalampos Skarlis
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Aikaterini Gkoufa
- Department of Infectious Diseases and COVID-19 Unit, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos V. Sipsas
- Department of Infectious Diseases and COVID-19 Unit, Laiko General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Clio P. Mavragani
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Fourth Department of Internal Medicine, School of Medicine, University Hospital Attikon, National and Kapodistrian University of Athens, Athens, Greece
- Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Athens, Greece
- *Correspondence: Clio P. Mavragani
| |
Collapse
|
13
|
Wei Q, Cai G, Chen G, Shen M, Gao Y, Ning X, Wang J, Jiang W. HIV infection is not associated with perioperative blood loss in patients undergoing total hip arthroplasty. J Orthop Surg Res 2022; 17:155. [PMID: 35264228 PMCID: PMC8905771 DOI: 10.1186/s13018-022-03055-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/03/2022] [Indexed: 11/17/2022] Open
Abstract
Background Patients with HIV have a higher prevalence of thrombocytopenia than those without HIV infection, increasing their risk of substantial perioperative blood loss (PBL) during total hip arthroplasty (THA). This study aimed to evaluate PBL risk factors in HIV-infected patients undergoing THA. Methods Eighteen HIV+ patients (21 hip joints) and 33 HIV− patients (36 joints) undergoing THA were enrolled in this study. PBL was calculated using the Gross equation, which comprises total blood loss (TBL), dominant blood loss (DBL), and hidden blood loss (HBL). Risk factors for post-THA PBL in both patient populations was evaluated using multivariable linear regression. Results At baseline, the HIV+ patients were younger, more likely to be male and to have elevated hemoglobin and albumin levels, and lower erythrocyte sedimentation rates than HIV− patients. There were no differences in the T-lymphocyte subsets or coagulation function between the two groups. Age and albumin level were identified as potential HBL risk factors after THA, and albumin level was associated with higher TBL. The unadjusted linear regression analysis showed that the HBL and TBL were significantly higher in HIV+ patients than in HIV− patients. However, after adjusting for other factors, no differences in DBL, HBL, or TBL were observed between HIV− and HIV+ patients. Conclusion PBL was similar in both groups undergoing THA, regardless of their HIV-infection status. THA surgery is a safe and effective procedure in HIV+ patients.
Collapse
Affiliation(s)
- Qifeng Wei
- Department of Orthopedics, The Third People's Hospital of Shenzhen, 29 Bulan Road, Shenzhen, 518112, Guangdong Province, China
| | - Gaorui Cai
- Department of Orthopedics, The Third People's Hospital of Shenzhen, 29 Bulan Road, Shenzhen, 518112, Guangdong Province, China
| | - Guoneng Chen
- Department of Orthopedics, The Third People's Hospital of Shenzhen, 29 Bulan Road, Shenzhen, 518112, Guangdong Province, China
| | - Maoye Shen
- Department of Orthopedics, The Third People's Hospital of Shenzhen, 29 Bulan Road, Shenzhen, 518112, Guangdong Province, China
| | - Ya Gao
- Department of Orthopedics, The Third People's Hospital of Shenzhen, 29 Bulan Road, Shenzhen, 518112, Guangdong Province, China
| | - Xianjia Ning
- Center of Clinical Epidemiology, The Third People's Hospital of Shenzhen, 29 Bulan Road, Shenzhen, 518112, Guangdong Province, China.,Laboratory of Epidemiology, Tianjin Neurological Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Jinghua Wang
- Center of Clinical Epidemiology, The Third People's Hospital of Shenzhen, 29 Bulan Road, Shenzhen, 518112, Guangdong Province, China. .,Laboratory of Epidemiology, Tianjin Neurological Institute, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Wenxue Jiang
- Department of Orthopedics, The Third People's Hospital of Shenzhen, 29 Bulan Road, Shenzhen, 518112, Guangdong Province, China.
| |
Collapse
|
14
|
Menter DG, Afshar-Kharghan V, Shen JP, Martch SL, Maitra A, Kopetz S, Honn KV, Sood AK. Of vascular defense, hemostasis, cancer, and platelet biology: an evolutionary perspective. Cancer Metastasis Rev 2022; 41:147-172. [PMID: 35022962 PMCID: PMC8754476 DOI: 10.1007/s10555-022-10019-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/04/2022] [Indexed: 01/08/2023]
Abstract
We have established considerable expertise in studying the role of platelets in cancer biology. From this expertise, we were keen to recognize the numerous venous-, arterial-, microvascular-, and macrovascular thrombotic events and immunologic disorders are caused by severe, acute-respiratory-syndrome coronavirus 2 (SARS-CoV-2) infections. With this offering, we explore the evolutionary connections that place platelets at the center of hemostasis, immunity, and adaptive phylogeny. Coevolutionary changes have also occurred in vertebrate viruses and their vertebrate hosts that reflect their respective evolutionary interactions. As mammals adapted from aquatic to terrestrial life and the heavy blood loss associated with placentalization-based live birth, platelets evolved phylogenetically from thrombocytes toward higher megakaryocyte-blebbing-based production rates and the lack of nuclei. With no nuclei and robust RNA synthesis, this adaptation may have influenced viral replication to become less efficient after virus particles are engulfed. Human platelets express numerous receptors that bind viral particles, which developed from archetypal origins to initiate aggregation and exocytic-release of thrombo-, immuno-, angiogenic-, growth-, and repair-stimulatory granule contents. Whether by direct, evolutionary, selective pressure, or not, these responses may help to contain virus spread, attract immune cells for eradication, and stimulate angiogenesis, growth, and wound repair after viral damage. Because mammalian and marsupial platelets became smaller and more plate-like their biophysical properties improved in function, which facilitated distribution near vessel walls in fluid-shear fields. This adaptation increased the probability that platelets could then interact with and engulf shedding virus particles. Platelets also generate circulating microvesicles that increase membrane surface-area encounters and mark viral targets. In order to match virus-production rates, billions of platelets are generated and turned over per day to continually provide active defenses and adaptation to suppress the spectrum of evolving threats like SARS-CoV-2.
Collapse
Affiliation(s)
- David G Menter
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Vahid Afshar-Kharghan
- Division of Internal Medicine, Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - John Paul Shen
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephanie L Martch
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Kopetz
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenneth V Honn
- Department of Pathology, Bioactive Lipids Research Program, Wayne State University, 5101 Cass Ave. 430 Chemistry, Detroit, MI, 48202, USA
- Department of Pathology, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA
- Cancer Biology Division, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA
| | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
15
|
Scheim DE. A Deadly Embrace: Hemagglutination Mediated by SARS-CoV-2 Spike Protein at Its 22 N-Glycosylation Sites, Red Blood Cell Surface Sialoglycoproteins, and Antibody. Int J Mol Sci 2022; 23:2558. [PMID: 35269703 PMCID: PMC8910562 DOI: 10.3390/ijms23052558] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/11/2022] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
Rouleaux (stacked clumps) of red blood cells (RBCs) observed in the blood of COVID-19 patients in three studies call attention to the properties of several enveloped virus strains dating back to seminal findings of the 1940s. For COVID-19, key such properties are: (1) SARS-CoV-2 binds to RBCs in vitro and also in the blood of COVID-19 patients; (2) although ACE2 is its target for viral fusion and replication, SARS-CoV-2 initially attaches to sialic acid (SA) terminal moieties on host cell membranes via glycans on its spike protein; (3) certain enveloped viruses express hemagglutinin esterase (HE), an enzyme that releases these glycan-mediated bindings to host cells, which is expressed among betacoronaviruses in the common cold strains but not the virulent strains, SARS-CoV, SARS-CoV-2 and MERS. The arrangement and chemical composition of the glycans at the 22 N-glycosylation sites of SARS-CoV-2 spike protein and those at the sialoglycoprotein coating of RBCs allow exploration of specifics as to how virally induced RBC clumping may form. The in vitro and clinical testing of these possibilities can be sharpened by the incorporation of an existing anti-COVID-19 therapeutic that has been found in silico to competitively bind to multiple glycans on SARS-CoV-2 spike protein.
Collapse
Affiliation(s)
- David E Scheim
- US Public Health Service, Commissioned Officer, Inactive Reserve, Blacksburg, VA 24060, USA
| |
Collapse
|
16
|
Ostermeier B, Soriano-Sarabia N, Maggirwar SB. Platelet-Released Factors: Their Role in Viral Disease and Applications for Extracellular Vesicle (EV) Therapy. Int J Mol Sci 2022; 23:2321. [PMID: 35216433 PMCID: PMC8876984 DOI: 10.3390/ijms23042321] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/13/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
Platelets, which are small anuclear cell fragments, play important roles in thrombosis and hemostasis, but also actively release factors that can both suppress and induce viral infections. Platelet-released factors include sCD40L, microvesicles (MVs), and alpha granules that have the capacity to exert either pro-inflammatory or anti-inflammatory effects depending on the virus. These factors are prime targets for use in extracellular vesicle (EV)-based therapy due to their ability to reduce viral infections and exert anti-inflammatory effects. While there are some studies regarding platelet microvesicle-based (PMV-based) therapy, there is still much to learn about PMVs before such therapy can be used. This review provides the background necessary to understand the roles of platelet-released factors, how these factors might be useful in PMV-based therapy, and a critical discussion of current knowledge of platelets and their role in viral diseases.
Collapse
Affiliation(s)
| | | | - Sanjay B. Maggirwar
- Department of Microbiology Immunology and Tropical Medicine, The George Washington University, 2300 I Street NW, Washington, DC 20037, USA; (B.O.); (N.S.-S.)
| |
Collapse
|
17
|
Platelets in COVID-19 disease: friend, foe, or both? Pharmacol Rep 2022; 74:1182-1197. [PMID: 36463349 PMCID: PMC9726679 DOI: 10.1007/s43440-022-00438-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 12/07/2022]
Abstract
Immuno-thrombosis of COVID-19 results in the activation of platelets and coagulopathy. Antiplatelet therapy has been widely used in COVID-19 patients to prevent thrombotic events. However, recent analysis of clinical trials does not support the major effects of antiplatelet therapy on mortality in hospitalized COVID-19 patients, despite the indisputable evidence for an increased risk of thrombotic complications in COVID-19 disease. This apparent paradox calls for an explanation. Platelets have an important role in sensing and orchestrating host response to infection, and several platelet functions related to host defense response not directly related to their well-known hemostatic function are emerging. In this paper, we aim to review the evidence supporting the notion that platelets have protective properties in maintaining endothelial barrier integrity in the course of an inflammatory response, and this role seems to be of particular importance in the lung. It might, thus, well be that the inhibition of platelet function, if affecting the protective aspect of platelet activity, might diminish clinical benefits resulting from the inhibition of the pro-thrombotic phenotype of platelets in immuno-thrombosis of COVID-19. A better understanding of the platelet-dependent mechanisms involved in the preservation of the endothelial barrier is necessary to design the antiplatelet therapeutic strategies that inhibit the pro-thrombotic activity of platelets without effects on the vaso-protective function of platelets safeguarding the pulmonary endothelial barrier during multicellular host defense in pulmonary circulation.
Collapse
|
18
|
Li T, Yang Y, Li Y, Wang Z, Ma F, Luo R, Xu X, Zhou G, Wang J, Niu J, Lv G, Crispe IN, Tu Z. Platelets mediate inflammatory monocyte activation by SARS-CoV-2 Spike protein. J Clin Invest 2021; 132:150101. [PMID: 34964720 PMCID: PMC8843740 DOI: 10.1172/jci150101] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 12/21/2021] [Indexed: 01/08/2023] Open
Abstract
Infection with SARS-CoV-2, the causative agent of COVID-19, causes mild to moderate disease in most patients but carries a risk of morbidity and mortality. Seriously affected individuals manifest disorders of hemostasis and a cytokine storm, but it is not understood how these manifestations of severe COVID-19 are linked. Here, we showed that the SARS-CoV-2 spike protein engaged the CD42b receptor to activate platelets via 2 distinct signaling pathways and promoted platelet-monocyte communication through the engagement of P selectin/PGSL-1 and CD40L/CD40, which led to proinflammatory cytokine production by monocytes. These results explain why hypercoagulation, monocyte activation, and a cytokine storm are correlated in patients severely affected by COVID-19 and suggest a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Tianyang Li
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yang Yang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yongqi Li
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhengmin Wang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Faxiang Ma
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Runqi Luo
- Department of Infectious Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaoming Xu
- Department of Infectious Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Guo Zhou
- Department of Infectious Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jianhua Wang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Junqi Niu
- Institute of Liver Diseases, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Guoyue Lv
- Institute of Liver Diseases, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ian N. Crispe
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Zhengkun Tu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
- Institute of Liver Diseases, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
19
|
Poveda-Jaramillo R. Coronavirus disease 2019-induced hypercoagulability and its clinical implications. Asian Cardiovasc Thorac Ann 2021; 30:515-523. [PMID: 34930050 DOI: 10.1177/02184923211069185] [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: 01/08/2023]
Abstract
Coronavirus disease 2019 is the disease produced by severe acute respiratory syndrome-coronavirus-2, which is introduced into the host's cell thanks to the angiotensin-converting enzyme 2 receptor. Once there, it uses the cell's machinery to multiply itself. In this process, it generates an immune response that stimulates the lymphocytes to produce cytokines and reactive oxygen species that begin to deteriorate the endothelial cell. Complement activation, through the complement attack complex and C5a, contributes to this endothelial damage. The different mediators further promote the expression of adhesion molecules on the endothelial surface, which encourages all blood cells to adhere to the endothelial surface to form small conglomerates, called clots, which obstruct the lumen of the small blood vessels. Furthermore, the mediators of clot lysis are inhibited. All this promotes a prothrombotic environment within the pulmonary capillaries that is reflected in the elevation of D-dimer. The only solution for this cascade of events seems to be the implementation of an effective anticoagulation protocol that early counteracts the changes induced by thrombi in the pulmonary circulation and reflected in the functioning of the right ventricle.
Collapse
|
20
|
Harun H, Nayoan CR, White I, Syamsi N, Tanra AAM, Sarifuddin S. Platelet Index in Patients with Confirmed Coronavirus Disease 2019 Cases. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.7743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Changes found on hematological examination are a helpful modality for assessing coronavirus disease 2019 (COVID-19). In addition, platelet index is a parameter that can help in assessing the COVID-19 disease prognosis.
AIM: Objective of the study is to determine the difference in platelet index in nucleid acid amplification test (NAAT) confirmed COVID-19 patients, suspected COVID-19 with negative NAAT results, and non-COVID-19 controls.
METHODS: This is an analytical observational study with 96 subjects; 48 subjects with confirmed COVID-19, 23 subjects suspected COVID-19 with negative NAAT results, and 24 non-COVID-19 control subjects. First, NAAT examination was carried out using the GeneXpert tool with the target genes of the E and N2 genes. Then, the platelet index was compared between the three groups by the Kruskal-Wallis test.
RESULTS: There was no significant difference in the number of platelet (PLT), mean PLT volume (MPV), and PLT crit (PCT) between the three groups with p = 0.732, 0.741 and 0.483, respectively. In general, the number of PLT, MPV, and PCT in the three groups was within the normal reference value.
CONCLUSIONS: There were no significant differences observed in the number of PLT, MPV, and PCT between COVID-19 patients with positive NAAT, COVID-19 suspects with negative NAAT, and non-COVID-19 controls. Therefore, detecting the severe acute respiratory syndrome coronavirus 2 virus by NAAT examination in COVID-19 patients has not altered the PLT index changes.
Collapse
|
21
|
Mencarini T, Roka-Moiia Y, Bozzi S, Redaelli A, Slepian MJ. Electrical impedance vs. light transmission aggregometry: Testing platelet reactivity to antiplatelet drugs using the MICELI POC impedance aggregometer as compared to a commercial predecessor. Thromb Res 2021; 204:66-75. [PMID: 34147831 PMCID: PMC11416791 DOI: 10.1016/j.thromres.2021.05.021] [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: 01/29/2021] [Revised: 05/08/2021] [Accepted: 05/26/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND Patients' responses to antiplatelet therapy significantly vary, with individuals showing high residual platelet reactivity associated with thrombosis. To personalize thrombosis management, platelet function testing has been suggested as a promising tool able to monitor the antithrombotic effect of antiplatelet agents in real-time. We have prototyped the MICELI, a miniature and easy-to-use electrical impedance aggregometer (EIA), measuring platelet aggregation in whole blood. Here, we tested the capability of the MICELI aggregometer to quantify platelet reactivity on antiplatelet agents, as compared with conventional light-transmission aggregometry (LTA). METHODS Platelet aggregation in ACD-anticoagulated whole blood and platelet-rich plasma of healthy donors (n = 30) was evaluated. The effect of clopidogrel, ticagrelor, cangrelor, cilostazol, and tirofiban on ADP-induced aggregation was tested, while aspirin was evaluated with arachidonic acid and collagen. Platelet aggregation was recorded using the MICELI or BioData PAP-8E (Bio/Data Corp.) aggregometers. RESULTS The MICELI aggregometer detected an adequate and comparable dose-dependent decrease of platelet aggregation in response to increments of drugs' concentrations, as compared to LTA (the inter-device R2 = 0.79-0.93). Platelet aggregation in platelet-rich plasma recorded by LTA showed higher sensitivity to antiplatelet agents, but it couldn't distinguish between different drug doses as indicated by saturation of the aggregatory response. CONCLUSION Platelet aggregation in whole blood as recorded by EIA represents a better model system for evaluation of platelet reactivity as compared with platelet aggregation in platelet-rich plasma as recorded by LTA, since EIA takes into consideration the modulatory effect of other blood cells on platelet hemostatic function and pharmacodynamics of antiplatelet drugs in vivo. As such, the MICELI impedance aggregometer could be potentially employed for the point-of-care monitoring of platelet function in patients on-treatment for personalized tailoring of their antiplatelet regimen.
Collapse
Affiliation(s)
- Tatiana Mencarini
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Yana Roka-Moiia
- Department of Medicine, Sarver Heart Center, University of Arizona, Tucson, AZ, United States of America; Department of Biomedical Engineering, Sarver Heart Center, University of Arizona, Tucson, AZ, United States of America
| | - Silvia Bozzi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Alberto Redaelli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Marvin J Slepian
- Department of Medicine, Sarver Heart Center, University of Arizona, Tucson, AZ, United States of America; Department of Biomedical Engineering, Sarver Heart Center, University of Arizona, Tucson, AZ, United States of America.
| |
Collapse
|
22
|
Hottz ED, Quirino-Teixeira AC, Merij LB, Pinheiro MBM, Rozini SV, Bozza FA, Bozza PT. Platelet-leukocyte interactions in the pathogenesis of viral infections. Platelets 2021; 33:200-207. [PMID: 34260328 DOI: 10.1080/09537104.2021.1952179] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Evolving evidence demonstrates that platelets have major roles in viral syndromes through previously unrecognized viral sensing and effector functions. Activated platelets and increased platelet-leukocyte aggregates are observed in clinical and experimental viral infections. The mechanisms and outcomes of platelet-leukocyte interactions depend on the interacting leukocyte as well as on the pathogen and pathological conditions. In this review, we discuss the mechanisms involved in platelet interactions with leukocytes and its functions during viral infections. We focus on the contributions of human platelet-leukocyte interactions to pathophysiological and protective responses during viral infections of major global health relevance, including acquired immunodeficiency syndrome (AIDS), dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS), influenza pneumonia, and COVID-19.
Collapse
Affiliation(s)
- Eugenio D Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil.,Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Anna Cecíllia Quirino-Teixeira
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Laura Botelho Merij
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Mariana Brandi Mendonça Pinheiro
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Stephane Vicente Rozini
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, MG, Brazil
| | - Fernando A Bozza
- Laboratory of Clinical Research in Intensive Care Medicine, National Institute of Infectious Disease Evandro Chagas, Oswaldo Cruz Foundation, Rio de Janeiro , Brazil.,Intensive Care Medicine, D'Or Institute for Research and Education, Rio de Janeiro, RJ, Brazil
| | - Patrícia T Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
23
|
Promoting platelets is a therapeutic option to combat severe viral infection of the lung. Blood Adv 2021; 4:1640-1642. [PMID: 32315397 PMCID: PMC7189284 DOI: 10.1182/bloodadvances.2020001669] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 03/25/2020] [Indexed: 02/07/2023] Open
|
24
|
Patel P, Michael JV, Naik UP, McKenzie SE. Platelet FcγRIIA in immunity and thrombosis: Adaptive immunothrombosis. J Thromb Haemost 2021; 19:1149-1160. [PMID: 33587783 DOI: 10.1111/jth.15265] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/14/2021] [Accepted: 02/09/2021] [Indexed: 12/16/2022]
Abstract
Sepsis and autoimmune diseases remain major causes of morbidity and mortality. The last decade has seen a new appreciation of platelets in host defense, in both immunity and thrombosis. Platelets are first responders in the blood to microbes or non-microbial antigens. The role of platelets in physiologic immunity is counterbalanced by their role in pathology, for example, microvascular thrombosis. Platelets encounter microbes and antigens via both innate and adaptive immune processes; platelets also help to shape the subsequent adaptive response. FcγRIIA is a receptor for immune complexes opsonized by IgG or pentraxins, and expressed in humans by platelets, granulocytes, monocytes and macrophages. With consideration of the roles of IgG and Fc receptors, the host response to microbes and autoantigens can be called adaptive immunothrombosis. Here we review newer developments involving platelet FcγRIIA in humans and humanized mice in immunity and thrombosis, with special attention to heparin-induced thrombocytopenia, systemic lupus erythematosus, and bacterial sepsis. Human genetic diversity in platelet receptors and the utility of humanized mouse models are highlighted.
Collapse
Affiliation(s)
- Pravin Patel
- Department of Medicine, Cardeza Foundation for Hematological Research, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - James V Michael
- Department of Medicine, Cardeza Foundation for Hematological Research, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ulhas P Naik
- Department of Medicine, Cardeza Foundation for Hematological Research, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Steven E McKenzie
- Department of Medicine, Cardeza Foundation for Hematological Research, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
25
|
Pretorius E. Platelets in HIV: A Guardian of Host Defence or Transient Reservoir of the Virus? Front Immunol 2021; 12:649465. [PMID: 33968041 PMCID: PMC8102774 DOI: 10.3389/fimmu.2021.649465] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/06/2021] [Indexed: 01/28/2023] Open
Abstract
The immune and inflammatory responses of platelets to human immunodeficiency virus 1 (HIV-1) and its envelope proteins are of great significance to both the treatment of the infection, and to the comorbidities related to systemic inflammation. Platelets can interact with the HIV-1 virus itself, or with viral membrane proteins, or with dysregulated inflammatory molecules in circulation, ensuing from HIV-1 infection. Platelets can facilitate the inhibition of HIV-1 infection via endogenously-produced inhibitors of HIV-1 replication, or the virus can temporarily hide from the immune system inside platelets, whereby platelets act as HIV-1 reservoirs. Platelets are therefore both guardians of the host defence system, and transient reservoirs of the virus. Such reservoirs may be of particular significance during combination antiretroviral therapy (cART) interruption, as it may drive viral persistence, and result in significant implications for treatment. Both HIV-1 envelope proteins and circulating inflammatory molecules can also initiate platelet complex formation with immune cells and erythrocytes. Complex formation cause platelet hypercoagulation and may lead to an increased thrombotic risk. Ultimately, HIV-1 infection can initiate platelet depletion and thrombocytopenia. Because of their relatively short lifespan, platelets are important signalling entities, and could be targeted more directly during HIV-1 infection and cART.
Collapse
Affiliation(s)
- Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
26
|
Abstract
Platelets play an essential role in maintaining vascular integrity after injury. In addition, platelets contribute to the immune response to pathogens. For instance, they express receptors that mediate binding of viruses, and toll-like receptors that activate the cell in response to pathogen-associated molecular patterns. Platelets can be beneficial and/or detrimental during viral infections. They reduce blood-borne viruses by engulfing the free virus and presenting the virus to neutrophils. However, platelets can also enhance inflammation and tissue injury during viral infections. Here, we discuss the roles of platelets in viral infection.
Collapse
Affiliation(s)
- Silvio Antoniak
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nigel Mackman
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
27
|
Gómez RM, López Ortiz AO, Schattner M. New roles of platelets in inflammation. CURRENT OPINION IN PHYSIOLOGY 2021. [DOI: 10.1016/j.cophys.2020.08.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
28
|
Neu CT, Gutschner T, Haemmerle M. Post-Transcriptional Expression Control in Platelet Biogenesis and Function. Int J Mol Sci 2020; 21:ijms21207614. [PMID: 33076269 PMCID: PMC7589263 DOI: 10.3390/ijms21207614] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 10/06/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
Platelets are highly abundant cell fragments of the peripheral blood that originate from megakaryocytes. Beside their well-known role in wound healing and hemostasis, they are emerging mediators of the immune response and implicated in a variety of pathophysiological conditions including cancer. Despite their anucleate nature, they harbor a diverse set of RNAs, which are subject to an active sorting mechanism from megakaryocytes into proplatelets and affect platelet biogenesis and function. However, sorting mechanisms are poorly understood, but RNA-binding proteins (RBPs) have been suggested to play a crucial role. Moreover, RBPs may regulate RNA translation and decay following platelet activation. In concert with other regulators, including microRNAs, long non-coding and circular RNAs, RBPs control multiple steps of the platelet life cycle. In this review, we will highlight the different RNA species within platelets and their impact on megakaryopoiesis, platelet biogenesis and platelet function. Additionally, we will focus on the currently known concepts of post-transcriptional control mechanisms important for RNA fate within platelets with a special emphasis on RBPs.
Collapse
Affiliation(s)
- Carolin T. Neu
- Institute of Pathology, Section for Experimental Pathology, Medical Faculty, Martin-Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany;
| | - Tony Gutschner
- Junior Research Group ‘RNA Biology and Pathogenesis’, Medical Faculty, Martin-Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany;
| | - Monika Haemmerle
- Institute of Pathology, Section for Experimental Pathology, Medical Faculty, Martin-Luther University Halle-Wittenberg, 06120 Halle/Saale, Germany;
- Correspondence: ; Tel.: +49-345-557-3964
| |
Collapse
|
29
|
He J, Wei Y, Chen J, Chen F, Gao W, Lu X. Dynamic trajectory of platelet-related indicators and survival of severe COVID-19 patients. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:607. [PMID: 33054834 PMCID: PMC7556573 DOI: 10.1186/s13054-020-03339-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 01/14/2023]
Affiliation(s)
- Jieyu He
- Department of Epidemiology and Biostatistics, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yongyue Wei
- Department of Epidemiology and Biostatistics, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jiao Chen
- Department of Geriatrics, Sir Run Run Hospital, Nanjing Medical University, 109 Longmian Avenue, Nanjing, 211166, China
| | - Feng Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China
| | - Wei Gao
- Department of Geriatrics, Sir Run Run Hospital, Nanjing Medical University, 109 Longmian Avenue, Nanjing, 211166, China.
| | - Xiang Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China. .,Department of Geriatrics, Sir Run Run Hospital, Nanjing Medical University, 109 Longmian Avenue, Nanjing, 211166, China.
| |
Collapse
|
30
|
Wool GD, Miller JL. The Impact of COVID-19 Disease on Platelets and Coagulation. Pathobiology 2020; 88:15-27. [PMID: 33049751 PMCID: PMC7649697 DOI: 10.1159/000512007] [Citation(s) in RCA: 266] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/05/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) causes a spectrum of disease; some patients develop a severe proinflammatory state which can be associated with a unique coagulopathy and procoagulant endothelial phenotype. Initially, COVID-19 infection produces a prominent elevation of fibrinogen and D-dimer/fibrin(ogen) degradation products. This is associated with systemic hypercoagulability and frequent venous thromboembolic events. The degree of D-dimer elevation positively correlates with mortality in COVID-19 patients. COVID-19 also leads to arterial thrombotic events (including strokes and ischemic limbs) as well as microvascular thrombotic disorders (as frequently documented at autopsy in the pulmonary vascular beds). COVID-19 patients often have mild thrombocytopenia and appear to have increased platelet consumption, together with a corresponding increase in platelet production. Disseminated intravascular coagulopathy (DIC) and severe bleeding events are uncommon in COVID-19 patients. Here, we review the current state of knowledge of COVID-19 and hemostasis.
Collapse
Affiliation(s)
- Geoffrey D Wool
- Department of Pathology, University of Chicago, Chicago, Illinois, USA,
| | - Jonathan L Miller
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
31
|
Feldman C, Anderson R. Platelets and Their Role in the Pathogenesis of Cardiovascular Events in Patients With Community-Acquired Pneumonia. Front Immunol 2020; 11:577303. [PMID: 33042161 PMCID: PMC7527494 DOI: 10.3389/fimmu.2020.577303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
Community-acquired pneumonia (CAP) remains an important cause of morbidity and mortality throughout the world with much recent and ongoing research focused on the occurrence of cardiovascular events (CVEs) during the infection, which are associated with adverse short-term and long-term survival. Much of the research directed at unraveling the pathogenesis of these events has been undertaken in the settings of experimental and clinical CAP caused by the dangerous, bacterial respiratory pathogen, Streptococcus pneumoniae (pneumococcus), which remains the most common bacterial cause of CAP. Studies of this type have revealed that although platelets play an important role in host defense against infection, there is also increasing recognition that hyperactivation of these cells contributes to a pro-inflammatory, prothrombotic systemic milieu that contributes to the etiology of CVEs. In the case of the pneumococcus, platelet-driven myocardial damage and dysfunction is exacerbated by the direct cardiotoxic actions of pneumolysin, a major pore-forming toxin of this pathogen, which also acts as potent activator of platelets. This review is focused on the role of platelets in host defense against infection, including pneumococcal infection in particular, and reviews the current literature describing the potential mechanisms by which platelet activation contributes to cardiovascular complications in CAP. This is preceded by an evaluation of the burden of pneumococcal infection in CAP, the clinical features and putative pathogenic mechanisms of the CVE, and concludes with an evaluation of the potential utility of the anti-platelet activity of macrolides and various adjunctive therapies.
Collapse
Affiliation(s)
- Charles Feldman
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, Institute of Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
32
|
Feldman C, Anderson R. Brief review: Cardiac complications and platelet activation in COVID-19 infection. Afr J Thorac Crit Care Med 2020; 26:10.7196/AJTCCM.2020.v26i3.107. [PMID: 34235425 PMCID: PMC7433708 DOI: 10.7196/ajtccm.2020.v26i3.107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2020] [Indexed: 12/19/2022] Open
Abstract
COVID-19 pneumonia, much like that of bacterial and viral community-acquired pneumonia before it, is accompanied by a high rate of cardio- and cerebrovascular events that are associated with an increased risk of complications and a greater mortality. Although the mechanisms underlying the pathogenesis of these adverse events are not entirely clear and may be multifactorial, platelets appear to have a prominent aetiologic role and this, together with an overview of the clinical evidence, forms the basis of this short review.
Collapse
Affiliation(s)
- C Feldman
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - R Anderson
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
33
|
Dib PRB, Quirino-Teixeira AC, Merij LB, Pinheiro MBM, Rozini SV, Andrade FB, Hottz ED. Innate immune receptors in platelets and platelet-leukocyte interactions. J Leukoc Biol 2020; 108:1157-1182. [PMID: 32779243 DOI: 10.1002/jlb.4mr0620-701r] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/11/2020] [Accepted: 06/28/2020] [Indexed: 12/14/2022] Open
Abstract
Platelets are chief cells in hemostasis. Apart from their hemostatic roles, platelets are major inflammatory effector cells that can influence both innate and adaptive immune responses. Activated platelets have thromboinflammatory functions linking hemostatic and immune responses in several physiological and pathological conditions. Among many ways in which platelets exert these functions, platelet expression of pattern recognition receptors (PRRs), including TLR, Nod-like receptor, and C-type lectin receptor families, plays major roles in sensing and responding to pathogen-associated or damage-associated molecular patterns (PAMPs and DAMPs, respectively). In this review, an increasing body of evidence is compiled showing the participation of platelet innate immune receptors, including PRRs, in infectious diseases, sterile inflammation, and cancer. How platelet recognition of endogenous DAMPs participates in sterile inflammatory diseases and thrombosis is discussed. In addition, platelet recognition of both PAMPs and DAMPs initiates platelet-mediated inflammation and vascular thrombosis in infectious diseases, including viral, bacterial, and parasite infections. The study also focuses on the involvement of innate immune receptors in platelet activation during cancer, and their contribution to tumor microenvironment development and metastasis. Finally, how innate immune receptors participate in platelet communication with leukocytes, modulating leukocyte-mediated inflammation and immune functions, is highlighted. These cell communication processes, including platelet-induced release of neutrophil extracellular traps, platelet Ag presentation to T-cells and platelet modulation of monocyte cytokine secretion are discussed in the context of infectious and sterile diseases of major concern in human health, including cardiovascular diseases, dengue, HIV infection, sepsis, and cancer.
Collapse
Affiliation(s)
- Paula Ribeiro Braga Dib
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil.,Laboratory of Immunology, Infectious Diseases and Obesity, Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Anna Cecíllia Quirino-Teixeira
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Laura Botelho Merij
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Mariana Brandi Mendonça Pinheiro
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Stephane Vicente Rozini
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Fernanda Brandi Andrade
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Eugenio Damaceno Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| |
Collapse
|
34
|
Banerjee M, Huang Y, Joshi S, Popa GJ, Mendenhall MD, Wang QJ, Garvy BA, Myint T, Whiteheart SW. Platelets Endocytose Viral Particles and Are Activated via TLR (Toll-Like Receptor) Signaling. Arterioscler Thromb Vasc Biol 2020; 40:1635-1650. [PMID: 32434410 PMCID: PMC7316618 DOI: 10.1161/atvbaha.120.314180] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Thrombocytopenia is associated with many viral infections suggesting virions interact with and affect platelets. Consistently, viral particles are seen inside platelets, and platelet activation markers are detected in viremic patients. In this article, we sought mechanistic insights into these virion/platelet interactions by examining how platelets endocytose, traffic, and are activated by a model virion. Approach and Results: Using fluorescently tagged HIV-1 pseudovirions, 3-dimensional structured illumination microscopy, and transgenic mouse models, we probed the interactions between platelets and virions. Mouse platelets used known endocytic machinery, that is, dynamin, VAMP (vesicle-associated membrane protein)-3, and Arf6 (ADP-ribosylation factor 6), to take up and traffic HIV-1 pseudovirions. Endocytosed HIV-1 pseudovirions trafficked through early (Rab4+) and late endosomes (Rab7+), and then to an LC3+ (microtubule-associated protein 1A/1B-light chain 3) compartment. Incubation with virions induced IRAK4 (interleukin 1 receptor-associated kinase 4), Akt (protein kinase B), and IKK (IκB kinase) activation, granule secretion, and platelet-leukocyte aggregate formation. This activation required TLRs (Toll-like receptors) and MyD88 (myeloid differentiation primary response protein 88) but was less extensive and slower than activation with thrombin. In vivo, HIV-1 pseudovirions injection led to virion uptake and platelet activation, as measured by IKK activation, platelet-leukocyte aggregate formation, and mild thrombocytopenia. All were decreased in VAMP-3-/- and, megakaryocyte/platelet-specific, Arf6-/- mice. Similar platelet activation profiles (increased platelet-leukocyte aggregates, plasma platelet factor 4, and phospho-IκBα) were detected in newly diagnosed and antiretroviral therapy-controlled HIV-1+ patients. CONCLUSIONS Collectively, our data provide mechanistic insights into the cell biology of how platelets endocytose and process virions. We propose a mechanism by which platelets sample the circulation and respond to potential pathogens that they take up.
Collapse
Affiliation(s)
- Meenakshi Banerjee
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
| | - Yunjie Huang
- Division of Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Smita Joshi
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
- Lexington VA Health Care System, Lexington, KY
| | - Gabriel J. Popa
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
| | - Michael D. Mendenhall
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
| | - Qing Jun Wang
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY
| | - Beth A. Garvy
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY
| | - Thein Myint
- Department of Infectious Diseases, Bluegrass Care Clinic, Kentucky Clinic, University of Kentucky, Lexington, KY
| | - Sidney W. Whiteheart
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
- Lexington VA Health Care System, Lexington, KY
| |
Collapse
|
35
|
Handtke S, Thiele T. Large and small platelets-(When) do they differ? J Thromb Haemost 2020; 18:1256-1267. [PMID: 32108994 DOI: 10.1111/jth.14788] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 02/06/2023]
Abstract
Platelets are most important in providing cellular hemostasis but also take part in inflammation and immune processes. Increased platelet size has been regarded as a feature describing a young and more reactive subpopulation until studies were published which questioned this concept. Moreover, changes of platelet size given by the mean platelet volume (MPV) were described for immune thrombocytopenia, cardiovascular disease, atherosclerosis, venous thromboembolism, chronic lung disease, sepsis, cancer-associated thrombosis, autoimmune disorders, and others. This review summarizes the literature on what is known about platelets with different size and describes controversies of studies with large and small platelets putting a focus on their thrombogenicity, age, and on the association of MPV with the mentioned diseases.
Collapse
Affiliation(s)
- Stefan Handtke
- Institut für Immunologie und Transfusionsmedizin, Abteilung Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Thomas Thiele
- Institut für Immunologie und Transfusionsmedizin, Abteilung Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
36
|
Page MJ, Pretorius E. A Champion of Host Defense: A Generic Large-Scale Cause for Platelet Dysfunction and Depletion in Infection. Semin Thromb Hemost 2020; 46:302-319. [PMID: 32279287 PMCID: PMC7339151 DOI: 10.1055/s-0040-1708827] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Thrombocytopenia is commonly associated with sepsis and infections, which in turn are characterized by a profound immune reaction to the invading pathogen. Platelets are one of the cellular entities that exert considerable immune, antibacterial, and antiviral actions, and are therefore active participants in the host response. Platelets are sensitive to surrounding inflammatory stimuli and contribute to the immune response by multiple mechanisms, including endowing the endothelium with a proinflammatory phenotype, enhancing and amplifying leukocyte recruitment and inflammation, promoting the effector functions of immune cells, and ensuring an optimal adaptive immune response. During infection, pathogens and their products influence the platelet response and can even be toxic. However, platelets are able to sense and engage bacteria and viruses to assist in their removal and destruction. Platelets greatly contribute to host defense by multiple mechanisms, including forming immune complexes and aggregates, shedding their granular content, and internalizing pathogens and subsequently being marked for removal. These processes, and the nature of platelet function in general, cause the platelet to be irreversibly consumed in the execution of its duty. An exaggerated systemic inflammatory response to infection can drive platelet dysfunction, where platelets are inappropriately activated and face immunological destruction. While thrombocytopenia may arise by condition-specific mechanisms that cause an imbalance between platelet production and removal, this review evaluates a generic large-scale mechanism for platelet depletion as a repercussion of its involvement at the nexus of responses to infection.
Collapse
Affiliation(s)
- Martin J Page
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
37
|
Real F, Capron C, Sennepin A, Arrigucci R, Zhu A, Sannier G, Zheng J, Xu L, Massé JM, Greffe S, Cazabat M, Donoso M, Delobel P, Izopet J, Eugenin E, Gennaro ML, Rouveix E, Cramer Bordé E, Bomsel M. Platelets from HIV-infected individuals on antiretroviral drug therapy with poor CD4+ T cell recovery can harbor replication-competent HIV despite viral suppression. Sci Transl Med 2020; 12:12/535/eaat6263. [DOI: 10.1126/scitranslmed.aat6263] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 07/23/2019] [Accepted: 10/01/2019] [Indexed: 12/24/2022]
Abstract
In addition to hemostasis, human platelets have several immune functions and interact with infectious pathogens including HIV in vitro. Here, we report that platelets from HIV-infected individuals on combined antiretroviral drug therapy (ART) with low blood CD4+ T cell counts (<350 cells/μl) contained replication-competent HIV despite viral suppression. In vitro, human platelets harboring HIV propagated the virus to macrophages, a process that could be prevented with the biologic abciximab, an anti–integrin αIIb/β3 Fab. Furthermore, in our cohort, 88% of HIV-infected individuals on ART with viral suppression and with platelets containing HIV were poor immunological responders with CD4+ T cell counts remaining below <350 cells/μl for more than one year. Our study suggests that platelets may be transient carriers of HIV and may provide an alternative pathway for HIV dissemination in HIV-infected individuals on ART with viral suppression and poor CD4+ T cell recovery.
Collapse
Affiliation(s)
- Fernando Real
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Université de Paris, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | | | - Alexis Sennepin
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Université de Paris, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Riccardo Arrigucci
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Aiwei Zhu
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Université de Paris, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Gérémy Sannier
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Université de Paris, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Jonathan Zheng
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Université de Paris, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Lin Xu
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Université de Paris, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Jean-Marc Massé
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
- Electron Microscopy Platform, Institut Cochin, Université de Paris, Paris, France
| | - Ségolène Greffe
- Department of Internal Medicine, Hôpital Ambroise Paré, Boulogne, France
| | - Michelle Cazabat
- CHU de Toulouse, Hôpital Purpan, Laboratoire de Virologie, Toulouse, France
| | - Maribel Donoso
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Pierre Delobel
- INSERM U1043, Toulouse, France
- Université Toulouse III Paul-Sabatier, Faculté de Médecine Toulouse-Purpan, Toulouse, France
- CHU de Toulouse, Hôpital Purpan, Service des Maladies Infectieuses et Tropicales, Toulouse, France
| | - Jacques Izopet
- CHU de Toulouse, Hôpital Purpan, Laboratoire de Virologie, Toulouse, France
- INSERM U1043, Toulouse, France
- Université Toulouse III Paul-Sabatier, Faculté de Médecine Toulouse-Purpan, Toulouse, France
| | - Eliseo Eugenin
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Maria Laura Gennaro
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Elisabeth Rouveix
- Department of Internal Medicine, Hôpital Ambroise Paré, Boulogne, France
| | - Elisabeth Cramer Bordé
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Université de Paris, Paris, France
- INSERM U1016, Paris, France
- Hôpital Ambroise Paré, Boulogne-Billancourt, France
| | - Morgane Bomsel
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Université de Paris, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| |
Collapse
|
38
|
Abstract
Platelets have a well-recognized role in hemostasis and thrombosis, and they are important amplifiers of inflammation and innate immune responses. The formation of DNA extracellular traps (ETs) is a complex cellular mechanism, which occurs in response to microbial infections and sterile inflammation, and results in the release of DNA complexed with histones and various granular proteins. ETs were first discovered in neutrophils (NETs); however, it is now accepted that other leukocytes, including eosinophils (EETs) and monocytes/macrophages (MoETs/METs), can also generate them. Moreover, several types of ETs have been described.Increasing evidence has demonstrated that platelets modulate the formation of ETs. This review summarizes recent findings about the physiopathological role of platelets in the formation of ETs during infection and future perspectives in the field.
Collapse
Affiliation(s)
- Ricardo M Gómez
- Laboratorio De Virus Animales, Instituto De Biotecnología Y Biología Molecular, CONICET-UNLP, La Plata, Argentina.,Global Viral Network, Baltimore, MD, USA
| | - Aída O López Ortiz
- Laboratorio De Virus Animales, Instituto De Biotecnología Y Biología Molecular, CONICET-UNLP, La Plata, Argentina.,Laboratorio De Trombosis Experimental, Instituto De Medicina Experimental, CONICET-ANM, Buenos Aires, Argentina
| | - Mirta Schattner
- Laboratorio De Trombosis Experimental, Instituto De Medicina Experimental, CONICET-ANM, Buenos Aires, Argentina
| |
Collapse
|
39
|
Jerez-Dolz D, Torramade-Moix S, Palomo M, Moreno-Castaño A, Lopez-Vilchez I, Hernandez R, Badimon JJ, Zafar MU, Diaz-Ricart M, Escolar G. Internalization of microparticles by platelets is partially mediated by toll-like receptor 4 and enhances platelet thrombogenicity. Atherosclerosis 2019; 294:17-24. [PMID: 31945614 DOI: 10.1016/j.atherosclerosis.2019.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/22/2019] [Accepted: 12/19/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND AIMS Circulating platelet microparticles (PMP) are the most abundant in bloodstream, are highly procoagulant and contribute to cross-talk with inflammatory cells. The aim of the present study was to investigate the interactions of PMP with platelets and explore the involvement of toll-like receptor 4 (TLR-4). METHODS PMP were separated by ultracentrifugation of expired platelet concentrates and added to: i) washed platelets, to confirm uptake, by flow cytometry and confocal and transmission electron microscopy, ii) platelet rich plasma (PRP), to assess changes in platelet function due to uptake by aggregometry in response to ADP; and iii) whole blood, to evaluate heterotypic aggregate (HA) formation by flow cytometry. Moreover, whole blood previously enriched with platelets with internalized PMP was used to explore modifications in thromboelastometry parameters (ROTEM). The inhibitory action of anti-TLR-4 was investigated. RESULTS Confocal and ultrastructural microscopy studies revealed PMP internalization by platelets. Flow cytometry showed PMP-platelet association (p < 0.01 vs controls, at different PMP dilutions). PMP, at 1/20 dilution, increased HA (p < 0.05 vs controls), the percentage of maximal platelet aggregation to ADP (p < 0.05 vs controls), and accelerated clotting and clot formation times (p < 0.05 vs controls). Incubation of platelets with anti-TLR-4 prior to exposure to PMP reduced PMP-platelet association (p < 0.05 vs absence of the antibody), prevented HA formation, reduced maximal platelet aggregation and normalized ROTEM parameters. CONCLUSIONS Platelets exhibit internalization ability towards their own PMP, a process that potentiates their thrombogenicity and is partially mediated by the innate immunity receptor TLR-4.
Collapse
Affiliation(s)
- Didac Jerez-Dolz
- Hematopathology, Pathological Anatomy, Hospital Clinic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Sergi Torramade-Moix
- Hematopathology, Pathological Anatomy, Hospital Clinic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Marta Palomo
- Hematopathology, Pathological Anatomy, Hospital Clinic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; Josep Carreras Leukaemia Research Institute, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain; Barcelona Endothelium Team, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain
| | - Ana Moreno-Castaño
- Hematopathology, Pathological Anatomy, Hospital Clinic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Irene Lopez-Vilchez
- Hematopathology, Pathological Anatomy, Hospital Clinic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Rosa Hernandez
- Hematopathology, Pathological Anatomy, Hospital Clinic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Juan Jose Badimon
- Atherothrombosis Research Unit, Icahn School of Medicine at Mount Sinai, New York, USA
| | - M Urooj Zafar
- Atherothrombosis Research Unit, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Maribel Diaz-Ricart
- Hematopathology, Pathological Anatomy, Hospital Clinic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; Barcelona Endothelium Team, Hospital Clinic/University of Barcelona Campus, Barcelona, Spain
| | - Gines Escolar
- Hematopathology, Pathological Anatomy, Hospital Clinic of Barcelona, Biomedical Diagnosis Centre (CDB), Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain; Atherothrombosis Research Unit, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
40
|
Cognasse F, Garraud O. Cytokines and related molecules, and adverse reactions related to platelet concentrate transfusions. Transfus Clin Biol 2019; 26:144-146. [PMID: 31327557 DOI: 10.1016/j.tracli.2019.06.324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 11/25/2022]
Abstract
Platelet transfusion is a safe process, but during or after the process the recipient may experience an adverse reaction and occasionally a serious adverse reaction (SAR). Platelet concentrate transfusion may be liable for significant absence of beneficial response. Danger may manifest clinically or biologically; in the latter case, manifestations are frequently an absence of the expected response to the blood component by the recipient. Blood platelets exert roles in inflammation, especially through the immunomodulator complex CD40/CD40L (sCD40L). In this review, we concentrate on the inflammatory potential of platelets and their participation to SARs in transfusion.
Collapse
Affiliation(s)
- F Cognasse
- The Rhône-Alpes-Auvergne Regional Branch of the French National Blood System EFS, 42000 Saint-Étienne, France.
| | - O Garraud
- EA3064, Faculty of Medicine, University of Lyon, 42023 Saint-Étienne, France; Palliative Care Unit, the Ruffec Hospital, 16700 Ruffec, France; Institut National de la Transfusion Sanguine, 75015 Paris, France
| |
Collapse
|
41
|
Cognasse F, Laradi S, Berthelot P, Bourlet T, Marotte H, Mismetti P, Garraud O, Hamzeh-Cognasse H. Platelet Inflammatory Response to Stress. Front Immunol 2019; 10:1478. [PMID: 31316518 PMCID: PMC6611140 DOI: 10.3389/fimmu.2019.01478] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/13/2019] [Indexed: 12/02/2022] Open
Abstract
Blood platelets play a central hemostatic role, (i) as they repair vascular epithelial damage, and (ii) they play immune defense roles, as they have the capacity to produce and secrete various cytokines, chemokines, and related products. Platelets sense and respond to local dangers (infectious or not). Platelets, therefore, mediate inflammation, express and use receptors to bind infectious pathogen moieties and endogenous ligands, among other components. Platelets contribute to effective pathogen clearance. Damage-associated molecular patterns (DAMPs) are danger signals released during inflammatory stress, such as burns, trauma and infection. Each pathogen is recognized by its specific molecular signature or pathogen-associated molecular pattern (PAMP). Recent data demonstrate that platelets have the capacity to sense external danger signals (DAMPs or PAMPs) differentially through a distinct type of pathogen recognition receptor (such as Toll-like receptors). Platelets regulate the innate immune response to pathogens and/or endogenous molecules, presenting several types of “danger” signals using a complete signalosome. Platelets, therefore, use complex tools to mediate a wide range of functions from danger sensing to tissue repair. Moreover, we noted that the secretory capacity of stored platelets over time and the development of stress lesions by platelets upon collection, processing, and storage are considered stress signals. The key message of this review is the “inflammatory response to stress” function of platelets in an infectious or non-infectious context.
Collapse
Affiliation(s)
- Fabrice Cognasse
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France.,GIMAP-EA3064, Université de Lyon, Saint-Étienne, France
| | - Sandrine Laradi
- Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France.,GIMAP-EA3064, Université de Lyon, Saint-Étienne, France
| | - Philippe Berthelot
- GIMAP-EA3064, Université de Lyon, Saint-Étienne, France.,Laboratoire des Agents Infectieux et d'Hygiène, CHU de Saint-Etienne, Saint-Étienne, France
| | - Thomas Bourlet
- GIMAP-EA3064, Université de Lyon, Saint-Étienne, France.,Laboratoire des Agents Infectieux et d'Hygiène, CHU de Saint-Etienne, Saint-Étienne, France
| | - Hubert Marotte
- SAINBIOSE, INSERM U1059, University of Lyon, Saint-Étienne, France.,Department of Rheumatology, University Hospital of Saint-Etienne, Saint-Étienne, France
| | - Patrick Mismetti
- SAINBIOSE, INSERM U1059, University of Lyon, Saint-Étienne, France.,Vascular and Therapeutic Medicine Department, Saint-Etienne University Hospital Center, Saint-Étienne, France
| | - Olivier Garraud
- GIMAP-EA3064, Université de Lyon, Saint-Étienne, France.,Institut National de Transfusion Sanguine, Paris, France
| | | |
Collapse
|
42
|
Nandi S, Sproul EP, Nellenbach K, Erb M, Gaffney L, Freytes DO, Brown AC. Platelet-like particles dynamically stiffen fibrin matrices and improve wound healing outcomes. Biomater Sci 2019; 7:669-682. [PMID: 30608063 PMCID: PMC6385160 DOI: 10.1039/c8bm01201f] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Native platelets perform several critical functions within the context of wound healing, including participating in initial hemostasis and interacting with fibrin at the wound site to induce clot retraction. Platelet depletion or dysfunction due to trauma or disease can inhibit robust wound healing responses. There has been a focus recently on developing synthetic, non-immunogenic platelet mimetic technologies for the purpose of augmenting hemostatic responses in cases of deficient native platelet functionality. Here we describe the application of synthetic platelet-like particles (PLPs), capable of recapitulating the deformable platelet body and fibrin specificity found in native platelets, to enhance healing outcomes. We first demonstrate PLPs mimic activated platelet morphology and induce fibrin clot retraction. During clot retraction, native platelets generate forces within a fibrin network to stiffen the fibrin matrix; therefore, we hypothesized that our PLPs will likewise be able to stiffen provisional fibrin matrices. Due to previous studies indicating that increased matrix stiffness is linked to increased cellular migration, we further hypothesize that PLP-mediated fibrin stiffening will enhance cell migration and improve healing outcomes within in vitro and in vivo models of wound healing. PLPs were found to enhance fibroblast migration in in vitro models of early wound healing and enhance healing outcomes in an in vivo murine model of wound healing. These studies demonstrate the utility of PLPs for enhancing wound repair and also provide insight into the role of native platelet-mediated clot retraction in wound healing.
Collapse
Affiliation(s)
- Seema Nandi
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Raleigh, NC 27695, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Kosulin K, Kernbichler S, Pichler H, Lawitschka A, Geyeregger R, Witt V, Lion T. Post-transplant Replication of Torque Teno Virus in Granulocytes. Front Microbiol 2018; 9:2956. [PMID: 30555452 PMCID: PMC6281686 DOI: 10.3389/fmicb.2018.02956] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/16/2018] [Indexed: 12/17/2022] Open
Abstract
Torque Teno virus (TTV) in humans is characterized by ubiquitous occurrence in peripheral blood (PB), without any related disease described to date. Several studies reported a significant increase of TTV plasma DNA levels in allogeneic transplant recipients, and suggested a correlation of elevated virus titers with immunosuppression and transplant-related complications. However, the site of viral replication in this setting has remained unclear. We have studied TTV in serial plasma specimens derived from 43 pediatric allogeneic hematopoietic stem cell transplantation (HSCT) recipients by RQ-PCR, and found increasing TTV-DNA levels in all patients post-transplant, with a peak around day +100 and maximum virus copy numbers reaching 4 × 10E9/ml. To assess whether the virus replicates in PB-cells, leukocyte subsets including granulocytes, monocytes, NK-cells, T- and B-lymphocytes were serially isolated by flow-sorting for TTV analysis in 19 patients. The virus was undetectable in most cell types, but was identified in granulocytes in all instances, revealing a median DNA copy number increase of 1.8 logs between days +30–100 post-transplant. Our data therefore provide evidence for TTV replication in granulocytes in this setting. In a control cohort of immunocompetent children and in HSCT recipients before day +30, TTV positivity in granulocytes was less common (33%), and the copy numbers were considerably lower. However, rising TTV replication about 2 weeks after granulocyte engraftment (>500 cells/μl) was observed suggesting that granulocyte recovery might be required for TTV expansion in severely immunosuppressed transplant recipients.
Collapse
Affiliation(s)
- Karin Kosulin
- Children's Cancer Research Institute, Vienna, Austria
| | | | | | | | | | - Volker Witt
- St. Anna Children's Hospital, Vienna, Austria
| | - Thomas Lion
- Children's Cancer Research Institute, Vienna, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
44
|
Seyoum M, Enawgaw B, Melku M. Human blood platelets and viruses: defense mechanism and role in the removal of viral pathogens. Thromb J 2018; 16:16. [PMID: 30026673 PMCID: PMC6048695 DOI: 10.1186/s12959-018-0170-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/18/2018] [Indexed: 12/19/2022] Open
Abstract
Platelets are small non-nucleated cell fragments and the second most abundant cell that play crucial role in managing vascular integrity and regulating hemostasis. Recent finding shows, beyond its hemostatic function platelets also play a main role in fighting against pathogen including viruses. With their receptors, platelet interacts with viral pathogen and this interaction between platelets and viral pathogens result in activation of platelets. Activated platelet releases different molecules that have antiviral activity including kinocidins and other platelet microbicidal peptides. In addition, activated platelet has antiviral role by different mechanism including; phagocytosis of viral pathogen, produce reactive oxygen species and interact with and activate other immune cells. In other side, antiplatelet treatments are one of defending mechanism of viral pathogen. This narrative review summarizes what is known regarding the role of human platelets in fighting viral pathogen.
Collapse
Affiliation(s)
- Masresha Seyoum
- University of Gondar hospital, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
- Department of Hematology & Immunohematology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Bamlaku Enawgaw
- Department of Hematology & Immunohematology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Mulugeta Melku
- Department of Hematology & Immunohematology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
45
|
Dengue Virus Induces the Release of sCD40L and Changes in Levels of Membranal CD42b and CD40L Molecules in Human Platelets. Viruses 2018; 10:v10070357. [PMID: 29976871 PMCID: PMC6071282 DOI: 10.3390/v10070357] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/28/2018] [Accepted: 06/30/2018] [Indexed: 02/07/2023] Open
Abstract
Platelets are considered as significant players in innate and adaptive immune responses. The adhesion molecules they express, including P-selectin, CD40L, and CD42b, facilitate interactions with many cellular effectors. Upon interacting with a pathogen, platelets rapidly express and enhance their adhesion molecules, and secrete cytokines and chemokines. A similar phenomenon occurs after exposure of platelets to thrombin, an agonist extensively used for in vitro activation of these cells. It was recently reported that the dengue virus not only interacts with platelets but possibly infects them, which triggers an increased expression of adhesion molecule P-selectin as well as secretion of IL-1β. In the present study, surface molecules of platelets like CD40L, CD42b, CD62P, and MHC class I were evaluated at 4 h of interaction with dengue virus serotype 2 (DENV-2), finding that DENV-2 induced a sharp rise in the membrane expression of all these molecules. At 2 and 4 h of DENV-2 stimulation of platelets, a significantly greater secretion of soluble CD40L (sCD40L) was found (versus basal levels) as well as cytokines such as GM-CSF, IL-6, IL-8, IL-10, and TNF-α. Compared to basal, DENV-2 elicited more than two-fold increase in these cytokines. Compared to the thrombin-induced response, the level generated by DENV-2 was much higher for GM-CSF, IL-6, and TNF-α. All these events induced by DENV end up in conspicuous morphological changes observed in platelets by confocal microscopy and transmission electron microscopy, very different from those elicited by thrombin in a more physiological scenery.
Collapse
|
46
|
Hottz ED, Bozza FA, Bozza PT. Platelets in Immune Response to Virus and Immunopathology of Viral Infections. Front Med (Lausanne) 2018; 5:121. [PMID: 29761104 PMCID: PMC5936789 DOI: 10.3389/fmed.2018.00121] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/12/2018] [Indexed: 01/04/2023] Open
Abstract
Platelets are essential effector cells in hemostasis. Aside from their role in coagulation, platelets are now recognized as major inflammatory cells with key roles in the innate and adaptive arms of the immune system. Activated platelets have key thromboinflammatory functions linking coagulation to immune responses in various infections, including in response to virus. Recent studies have revealed that platelets exhibit several pattern recognition receptors (PRR) including those from the toll-like receptor, NOD-like receptor, and C-type lectin receptor family and are first-line sentinels in detecting and responding to pathogens in the vasculature. Here, we review the main mechanisms of platelets interaction with viruses, including their ability to sustain viral infection and replication, their expression of specialized PRR, and activation of thromboinflammatory responses against viruses. Finally, we discuss the role of platelet-derived mediators and platelet interaction with vascular and immune cells in protective and pathophysiologic responses to dengue, influenza, and human immunodeficiency virus 1 infections.
Collapse
Affiliation(s)
- Eugenio D Hottz
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.,Departamento de Bioquimica, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | - Fernando A Bozza
- Laboratório de Medicina Intensiva, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.,Instituto D'Or de Pesquisa e Ensino, Rio de Janeiro, Brazil
| | - Patrícia T Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
47
|
Thakur A, Qureshi A, Kumar M. vhfRNAi: a web-platform for analysis of host genes involved in viral infections discovered by genome wide RNAi screens. MOLECULAR BIOSYSTEMS 2018; 13:1377-1387. [PMID: 28561835 DOI: 10.1039/c6mb00841k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Knockdown of host genes using high-throughput genome-wide RNA interference screens has identified numerous host factors that affect viral infections, which would be helpful in understanding host-virus interactions. We have developed a vhfRNAi web resource based on genome-wide RNAi experiments for viruses. It contains experimental details of 12 249 entries (host factors + restriction factors) for 18 viruses. Simultaneously, this resource encompasses analysis of overlapping genes, genome wide association studies, gene ontology (GO), pathogen interacting proteins, interaction networks and pathway enrichment. Using overlap analysis, it was found that Influenza A virus shared overlapping host genes with the majority of viruses including Hepatitis C virus and Dengue virus 2. In the genome wide association studies analysis, 429 diseases/traits were mapped, of which obesity-related traits were the most common. GO analysis revealed that the major categories belonged to metabolic processes, molecule transport, signal transduction, proteolysis, etc. In the pathogen interacting protein analysis, protein interaction data from different resources can be explored for further understanding of host-virus biology. By pathway enrichment analysis, a total of 8955 genes were mapped on 303 pathways with most of the hits coming from metabolic pathways. We have found 491 genes that are not essential for the host but essential for the virus and can be targeted to inhibit the virus. These may be explored as potential candidates for drug targets. The resource is freely accessible at and will be useful in understanding host-virus biology as well as identification of targets for the development of antiviral therapeutics.
Collapse
Affiliation(s)
- Anamika Thakur
- Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research, Sector 39-A, Chandigarh-160036, India.
| | | | | |
Collapse
|
48
|
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. [PMID: 29533683 DOI: 10.1080/09537104.2018.1445842] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [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
|
49
|
Kullaya VI, de Mast Q, van der Ven A, elMoussaoui H, Kibiki G, Simonetti E, de Jonge MI, Ferwerda G. Platelets Modulate Innate Immune Response Against Human Respiratory Syncytial Virus In Vitro. Viral Immunol 2017; 30:576-581. [PMID: 28783457 DOI: 10.1089/vim.2016.0161] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Detection of respiratory syncytial virus (RSV) in blood, including mononuclear leukocytes and organs other than the lung, suggests that RSV disseminates outside the respiratory tract. In this study, the role of platelets in host defense against RSV was explored using an in vitro model. Platelets, also produced in the lungs, are increasingly recognized as an important part of host immune responses and may therefore play a role in modulating lung infections and clearing RSV viremia. In human peripheral blood mononuclear cells (PBMCs), platelets significantly reduced RSV infection of monocytes, monocyte activation, and interferon (IFN)α/γ production. Direct contact of platelets with PBMCs modulated the immune response when stimulated with Poly I:C (TLR3) and R848 (TLR7/8), Toll-like receptors (TLRs) involved in the recognition of RSV, and led to an enhanced IFNα/γ production. This suggested that reduction in RSV infection of monocytes in the presence of platelets could be IFN dependent; blocking IFNα receptor 2 (IFNAR2) on PBMCs indeed increased RSV infection. In addition, IFNs were not detected when PBMCs were stimulated with inactivated RSV, indicating that infection of monocytes was important for the induction of IFN responses and that the platelet-mediated reduced RSV infection was responsible for the decreased IFN levels. Furthermore, platelets could internalize RSV reducing the amount of viral particles that could infect monocytes. Our findings suggest that platelets may play a role in the clearance of RSV viremia by internalizing viral particles and by enhancing type I IFN production from PBMCs, which subsequently exert antiviral effect on host cells.
Collapse
Affiliation(s)
- Vesla I Kullaya
- 1 Department of Internal Medicine, Radboud University Medical Center , Nijmegen, the Netherlands .,2 Kilimanjaro Clinical Research Institute , Kilimanjaro Christian Medical Center, Moshi, Tanzania
| | - Quirijn de Mast
- 1 Department of Internal Medicine, Radboud University Medical Center , Nijmegen, the Netherlands
| | - Andre van der Ven
- 1 Department of Internal Medicine, Radboud University Medical Center , Nijmegen, the Netherlands
| | - Hicham elMoussaoui
- 3 Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud University Medical Center , Radboud Center for Infectious Diseases, Nijmegen, the Netherlands
| | - Gibson Kibiki
- 2 Kilimanjaro Clinical Research Institute , Kilimanjaro Christian Medical Center, Moshi, Tanzania
| | - Elles Simonetti
- 3 Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud University Medical Center , Radboud Center for Infectious Diseases, Nijmegen, the Netherlands
| | - Marien I de Jonge
- 3 Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud University Medical Center , Radboud Center for Infectious Diseases, Nijmegen, the Netherlands
| | - Gerben Ferwerda
- 3 Laboratory of Pediatric Infectious Diseases, Department of Pediatrics, Radboud University Medical Center , Radboud Center for Infectious Diseases, Nijmegen, the Netherlands
| |
Collapse
|
50
|
Acetylsalicylic acid differentially limits the activation and expression of cell death markers in human platelets exposed to Staphylococcus aureus strains. Sci Rep 2017; 7:5610. [PMID: 28717145 PMCID: PMC5514152 DOI: 10.1038/s41598-017-06024-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 06/07/2017] [Indexed: 12/14/2022] Open
Abstract
Beyond their hemostatic functions, platelets alter their inflammatory response according to the bacterial stimulus. Staphylococcus aureus is associated with exacerbated inflammation and thrombocytopenia, which is associated with poor prognosis during sepsis. Acetylsalicylic acid and statins prevent platelet aggregation and decrease the mortality rate during sepsis. Therefore, we assessed whether these two molecules could reduce in vitro platelet activation and the inflammatory response to S. aureus. Platelets were exposed to clinical strains of S. aureus in the presence or absence of acetylsalicylic acid or fluvastatin. Platelet activation, aggregation, and release of soluble sCD62P, sCD40 Ligand, RANTES and GROα were assessed. Platelet cell death was evaluated by analyzing the mitochondrial membrane potential, phosphatidylserine exposure, platelet microparticle release and caspase-3 activation. All S. aureus strains induced platelet activation but not aggregation and decreased the platelet count, the expression of cell death markers and the release of RANTES and GROα. Acetylsalicylic acid but not fluvastatin limited platelet activation and inflammatory factor release and restored the platelet count by protecting platelets from Staphylococcus-induced expression of cell death markers. This study demonstrates that acetylsalicylic acid limits S. aureus-induced effects on platelets by reducing cell death, revealing new strategies to reduce the platelet contribution to bacteremia-associated inflammation.
Collapse
|