101
|
Gut microbial metabolites as multi-kingdom intermediates. Nat Rev Microbiol 2020; 19:77-94. [PMID: 32968241 DOI: 10.1038/s41579-020-0438-4] [Citation(s) in RCA: 614] [Impact Index Per Article: 153.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2020] [Indexed: 02/07/2023]
Abstract
The gut microbiota contributes to host physiology through the production of a myriad of metabolites. These metabolites exert their effects within the host as signalling molecules and substrates for metabolic reactions. Although the study of host-microbiota interactions remains challenging due to the high degree of crosstalk both within and between kingdoms, metabolite-focused research has identified multiple actionable microbial targets that are relevant for host health. Metabolites, as the functional output of combined host and microorganism interactions, provide a snapshot in time of an extraordinarily complex multi-organism system. Although substantial work remains towards understanding host-microbiota interactions and the underlying mechanisms, we review the current state of knowledge for each of the major classes of microbial metabolites with emphasis on clinical and translational research implications. We provide an overview of methodologies available for measurement of microbial metabolites, and in addition to discussion of key challenges, we provide a potential framework for integration of discovery-based metabolite studies with mechanistic work. Finally, we highlight examples in the literature where this approach has led to substantial progress in understanding host-microbiota interactions.
Collapse
|
102
|
Huot S, Laflamme C, Fortin PR, Boilard E, Pouliot M. IgG-aggregates rapidly upregulate FcgRI expression at the surface of human neutrophils in a FcgRII-dependent fashion: A crucial role for FcgRI in the generation of reactive oxygen species. FASEB J 2020; 34:15208-15221. [PMID: 32946139 DOI: 10.1096/fj.202001085r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/26/2020] [Accepted: 09/02/2020] [Indexed: 01/17/2023]
Abstract
Autoimmune complexes are an important feature of several autoimmune diseases such as lupus, as they contribute to tissue damage through the activation of immune cells. Neutrophils, key players in lupus, interact with immune complexes through Fc gamma receptors (FcgR). Incubation of neutrophils with aggregated-IgGs caused degranulation and increased the surface expression of FcgRI within minutes in a concentration-dependent fashion. After 30 minutes, IgG aggregates (1 mg/mL) upregulated FcgRI by 4.95 ± 0.45-fold. FcgRI-positive neutrophils reached 67.24% ± 6.88% on HA-IgGs stimulated neutrophils, from 3.12% ± 1.62% in non-stimulated cells, ranking IgG-aggregates among the most potent known agonists. FcgRIIa, and possibly FcgRIIIa, appeared to mediate this upregulation. Also, FcgRI-dependent signaling proved necessary for reactive oxygen species (ROS) production in response to IgG-aggregates. Finally, combinations of bacterial materials with aggregates dramatically boosted ROS production. This work suggests FcgRI as an essential component in the response of human neutrophils to immune complexes leading to the production of ROS, which may help explain how neutrophils contribute to tissue damage associated with immune complex-associated diseases, such as lupus.
Collapse
Affiliation(s)
- Sandrine Huot
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada.,Axe maladies infectieuses et immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Cynthia Laflamme
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada.,Axe maladies infectieuses et immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Paul R Fortin
- Axe maladies infectieuses et immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada.,Division de Rhumatologie, Département de Médecine, CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Eric Boilard
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada.,Axe maladies infectieuses et immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Marc Pouliot
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada.,Axe maladies infectieuses et immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| |
Collapse
|
103
|
Maouia A, Rebetz J, Kapur R, Semple JW. The Immune Nature of Platelets Revisited. Transfus Med Rev 2020; 34:209-220. [PMID: 33051111 PMCID: PMC7501063 DOI: 10.1016/j.tmrv.2020.09.005] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 01/08/2023]
Abstract
Platelets are the primary cellular mediators of hemostasis and this function firmly acquaints them with a variety of inflammatory processes. For example, platelets can act as circulating sentinels by expressing Toll-like receptors (TLR) that bind pathogens and this allows platelets to effectively kill them or present them to cells of the immune system. Furthermore, activated platelets secrete and express many pro- and anti-inflammatory molecules that attract and capture circulating leukocytes and direct them to inflamed tissues. In addition, platelets can directly influence adaptive immune responses via secretion of, for example, CD40 and CD40L molecules. Platelets are also the source of most of the microvesicles in the circulation and these miniscule elements further enhance the platelet’s ability to communicate with the immune system. More recently, it has been demonstrated that platelets and their parent cells, the megakaryocytes (MK), can also uptake, process and present both foreign and self-antigens to CD8+ T-cells conferring on them the ability to directly alter adaptive immune responses. This review will highlight several of the non-hemostatic attributes of platelets that clearly and rightfully place them as integral players in immune reactions. Platelets can act as circulating sentinels by expressing pathogen-associated molecular pattern receptors that bind pathogens and induce their killing and elimination. Activated platelets secrete and express a multitude of pro- and anti-inflammatory molecules that attract and capture circulating leukocytes and direct them to inflamed tissues. Platelets express and secrete many critical immunoregulatory molecules that significantly affect both innate and adaptive immune responses. Platelets are the primary source of microparticles in the circulation and these augment the platelet’s ability to communicate with the immune system. Platelets and megakaryocytes can act as antigen presenting cells and present both foreign- and self-peptides to T-cells.
Collapse
Affiliation(s)
- Amal Maouia
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Johan Rebetz
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Rick Kapur
- Sanquin Research, Department of Experimental Immunohematology, Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - John W Semple
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden; Clinical Immunology and Transfusion Medicine, Office of Medical Services, Region Skåne, Lund, Sweden.
| |
Collapse
|
104
|
Zaid Y, Puhm F, Allaeys I, Naya A, Oudghiri M, Khalki L, Limami Y, Zaid N, Sadki K, Ben El Haj R, Mahir W, Belayachi L, Belefquih B, Benouda A, Cheikh A, Langlois MA, Cherrah Y, Flamand L, Guessous F, Boilard E. Platelets Can Associate with SARS-Cov-2 RNA and Are Hyperactivated in COVID-19. Circ Res 2020; 127:1404-1418. [PMID: 32938299 PMCID: PMC7641188 DOI: 10.1161/circresaha.120.317703] [Citation(s) in RCA: 349] [Impact Index Per Article: 87.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022]
Abstract
Rationale: In addition to the overwhelming lung inflammation that prevails in COVID-19, hypercoagulation and thrombosis contribute to the lethality of subjects infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Platelets are chiefly implicated in thrombosis. Moreover, they can interact with viruses and are an important source of inflammatory mediators. While a lower platelet count is associated with severity and mortality, little is known about platelet function during COVID-19. Objective: To evaluate the contribution of platelets to inflammation and thrombosis in COVID-19 patients. Methods and Results: Blood was collected from 115 consecutive COVID-19 patients presenting non-severe (n=71) and severe (n=44) respiratory symptoms. We document the presence of SARS-CoV-2 RNA associated with platelets of COVID-19 patients. Exhaustive assessment of cytokines in plasma and in platelets revealed the modulation of platelet-associated cytokine levels in both non-severe and severe COVID-19 patients, pointing to a direct contribution of platelets to the plasmatic cytokine load. Moreover, we demonstrate that platelets release their alpha- and dense-granule contents in both non-severe and severe forms of COVID-19. In comparison to concentrations measured in healthy volunteers, phosphatidylserine-exposing platelet extracellular vesicles were increased in non-severe, but not in severe cases of COVID-19. Levels of D-dimers, a marker of thrombosis, failed to correlate with any measured indicators of platelet activation. Functionally, platelets were hyperactivated in COVID-19 subjects presenting non-severe and severe symptoms, with aggregation occurring at suboptimal thrombin concentrations. Furthermore, platelets adhered more efficiently onto collagen-coated surfaces under flow conditions. Conclusions: Taken together, the data suggest that platelets are at the frontline of COVID-19 pathogenesis, as they release various sets of molecules through the different stages of the disease. Platelets may thus have the potential to contribute to the overwhelming thrombo-inflammation in COVID-19, and the inhibition of pathways related to platelet activation may improve the outcomes during COVID-19.
Collapse
Affiliation(s)
- Younes Zaid
- Research Center of Abulcasis University of Health Sciences, Cheikh Zaïd Hospital, Rabat, Morocco (Y.Z., Y.L., R.B.E.H., W.M., L.B., B.B., A.B., A.C., Y.C.)
- Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco (Y.Z., N.Z., K.S.)
- Immunology and Biodiversity Laboratory, Biology, Faculty of Sciences Ain Chock, Hassan II University, Casablanca, Morocco (Y.Z., A.N., M.O., Y.L.)
| | - Florian Puhm
- Centre de Recherche du Centre Hospitalier Universitaire de Québec- Université Laval, Canada (F.P., I.A., L.F., E.B.)
- Département de microbiologie-infectiologie et d’immunologie, Université Laval, QC, Canada (F.P., I.A., L.F., E.B.)
| | - Isabelle Allaeys
- Centre de Recherche du Centre Hospitalier Universitaire de Québec- Université Laval, Canada (F.P., I.A., L.F., E.B.)
- Département de microbiologie-infectiologie et d’immunologie, Université Laval, QC, Canada (F.P., I.A., L.F., E.B.)
| | - Abdallah Naya
- Immunology and Biodiversity Laboratory, Biology, Faculty of Sciences Ain Chock, Hassan II University, Casablanca, Morocco (Y.Z., A.N., M.O., Y.L.)
| | - Mounia Oudghiri
- Immunology and Biodiversity Laboratory, Biology, Faculty of Sciences Ain Chock, Hassan II University, Casablanca, Morocco (Y.Z., A.N., M.O., Y.L.)
| | - Loubna Khalki
- Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco (L.K., F.G.)
| | - Youness Limami
- Research Center of Abulcasis University of Health Sciences, Cheikh Zaïd Hospital, Rabat, Morocco (Y.Z., Y.L., R.B.E.H., W.M., L.B., B.B., A.B., A.C., Y.C.)
- Immunology and Biodiversity Laboratory, Biology, Faculty of Sciences Ain Chock, Hassan II University, Casablanca, Morocco (Y.Z., A.N., M.O., Y.L.)
| | - Nabil Zaid
- Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco (Y.Z., N.Z., K.S.)
| | - Khalid Sadki
- Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco (Y.Z., N.Z., K.S.)
| | - Rafiqua Ben El Haj
- Research Center of Abulcasis University of Health Sciences, Cheikh Zaïd Hospital, Rabat, Morocco (Y.Z., Y.L., R.B.E.H., W.M., L.B., B.B., A.B., A.C., Y.C.)
| | - Wissal Mahir
- Research Center of Abulcasis University of Health Sciences, Cheikh Zaïd Hospital, Rabat, Morocco (Y.Z., Y.L., R.B.E.H., W.M., L.B., B.B., A.B., A.C., Y.C.)
| | - Lamiae Belayachi
- Research Center of Abulcasis University of Health Sciences, Cheikh Zaïd Hospital, Rabat, Morocco (Y.Z., Y.L., R.B.E.H., W.M., L.B., B.B., A.B., A.C., Y.C.)
| | - Bouchra Belefquih
- Research Center of Abulcasis University of Health Sciences, Cheikh Zaïd Hospital, Rabat, Morocco (Y.Z., Y.L., R.B.E.H., W.M., L.B., B.B., A.B., A.C., Y.C.)
| | - Amina Benouda
- Research Center of Abulcasis University of Health Sciences, Cheikh Zaïd Hospital, Rabat, Morocco (Y.Z., Y.L., R.B.E.H., W.M., L.B., B.B., A.B., A.C., Y.C.)
| | - Amine Cheikh
- Research Center of Abulcasis University of Health Sciences, Cheikh Zaïd Hospital, Rabat, Morocco (Y.Z., Y.L., R.B.E.H., W.M., L.B., B.B., A.B., A.C., Y.C.)
| | - Marc-André Langlois
- Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.L.)
| | - Yahia Cherrah
- Research Center of Abulcasis University of Health Sciences, Cheikh Zaïd Hospital, Rabat, Morocco (Y.Z., Y.L., R.B.E.H., W.M., L.B., B.B., A.B., A.C., Y.C.)
| | - Louis Flamand
- Centre de Recherche du Centre Hospitalier Universitaire de Québec- Université Laval, Canada (F.P., I.A., L.F., E.B.)
- Département de microbiologie-infectiologie et d’immunologie, Université Laval, QC, Canada (F.P., I.A., L.F., E.B.)
| | - Fadila Guessous
- Faculty of Medicine, Mohammed VI University of Health Sciences (UM6SS), Casablanca, Morocco (L.K., F.G.)
- Microbiology, Immunology and Cancer Biology, School of Medicine, University of Virginia, Charlottesville (F.G.)
| | - Eric Boilard
- Centre de Recherche du Centre Hospitalier Universitaire de Québec- Université Laval, Canada (F.P., I.A., L.F., E.B.)
- Département de microbiologie-infectiologie et d’immunologie, Université Laval, QC, Canada (F.P., I.A., L.F., E.B.)
| |
Collapse
|
105
|
CXCR4 hi effector neutrophils in sickle cell anemia: potential role for elevated circulating serotonin (5-HT) in CXCR4 hi neutrophil polarization. Sci Rep 2020; 10:14262. [PMID: 32868775 PMCID: PMC7459317 DOI: 10.1038/s41598-020-71078-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 08/07/2020] [Indexed: 01/01/2023] Open
Abstract
Leukocyte recruitment and heterocellular aggregate formation drive the inflammatory vaso-occlusive processes associated with sickle cell anemia (SCA). We characterized neutrophils in a population of patients with SCA and investigated whether platelet-derived molecules can induce phenotypic alterations in this cell type. Imaging flow cytometry analysis demonstrated that the frequency of circulating CXCR4hi neutrophils was significantly higher in steady-state SCA individuals than in healthy control individuals and that these cells presented increased CD11b activation and toll-like receptor-4 expression. SCA neutrophils display increased neutrophil-platelet aggregation, and CXCR4hi neutrophils demonstrated augmented neutrophil-platelet aggregate frequency with a higher mean number of platelets adhered per neutrophil. Importantly, incubation of neutrophils with platelets significantly elevated their CXCR4 expression, while SCA plasma was found to induce CXCR4hi neutrophil polarization significantly more than control plasma. SCA individuals had significantly increased plasma levels of serotonin (5-HT), and serotonin molecule and SCA plasma induced neutrophil CXCR4 expression in a serotonin-receptor-dependent manner. Thus, the augmented CXCR4hi neutrophil population may contribute to mechanisms that promote vaso-occlusion in SCA; furthermore, circulating serotonin, derived from platelet activation, may play a role in the polarization of neutrophils, suggesting that serotonin-receptor antagonists or serotonin reuptake inhibitors could represent therapeutic approaches to reduce neutrophil activation in SCA.
Collapse
|
106
|
Platelets Promote Macrophage Polarization toward Pro-inflammatory Phenotype and Increase Survival of Septic Mice. Cell Rep 2020; 28:896-908.e5. [PMID: 31340152 DOI: 10.1016/j.celrep.2019.06.062] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 02/28/2019] [Accepted: 06/17/2019] [Indexed: 12/21/2022] Open
Abstract
We investigated the contribution of human platelets to macrophage effector properties in the presence of lipopolysaccharide (LPS), as well as the beneficial effects and time frame for platelet transfusion in septic animals. Our results show that platelets sequester both pro-(TNF-α/IL-6) and anti-(IL-10) inflammatory cytokines released by monocytes. Low LPS concentrations (0.01 ng/mL) induced M2 macrophage polarization by decreasing CD64 and augmenting CD206 and CD163 expression; yet, the presence of platelets skewed monocytes toward type 1 macrophage (M1) phenotype in a cell-contact-dependent manner by the glycoprotein Ib (GPIb)-CD11b axis. Accordingly, platelet-licensed macrophages showed increased TNF-α levels, bacterial phagocytic activity, and a reduced healing capability. Platelet transfusion increased inducible nitric oxide synthase (iNOS)+ macrophages, improving bacterial clearance and survival rates in septic mice up to 6 h post-infection, an effect that was abolished by CD11b and GPIb blockade. Our results demonstrate that platelets orchestrate macrophage effector responses, improving the clinical outcome of sepsis in a narrow but relevant time frame.
Collapse
|
107
|
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
|
108
|
Abstract
The recent emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the ensuing global pandemic has presented a health emergency of unprecedented magnitude. Recent clinical data has highlighted that coronavirus disease 2019 (COVID-19) is associated with a significant risk of thrombotic complications ranging from microvascular thrombosis, venous thromboembolic disease, and stroke. Importantly, thrombotic complications are markers of severe COVID-19 and are associated with multiorgan failure and increased mortality. The evidence to date supports the concept that the thrombotic manifestations of severe COVID-19 are due to the ability of SARS-CoV-2 to invade endothelial cells via ACE-2 (angiotensin-converting enzyme 2), which is expressed on the endothelial cell surface. However, in patients with COVID-19 the subsequent endothelial inflammation, complement activation, thrombin generation, platelet, and leukocyte recruitment, and the initiation of innate and adaptive immune responses culminate in immunothrombosis, ultimately causing (micro)thrombotic complications, such as deep vein thrombosis, pulmonary embolism, and stroke. Accordingly, the activation of coagulation (eg, as measured with plasma D-dimer) and thrombocytopenia have emerged as prognostic markers in COVID-19. Given thrombotic complications are central determinants of the high mortality rate in COVID-19, strategies to prevent thrombosis are of critical importance. Several antithrombotic drugs have been proposed as potential therapies to prevent COVID-19-associated thrombosis, including heparin, FXII inhibitors, fibrinolytic drugs, nafamostat, and dipyridamole, many of which also possess pleiotropic anti-inflammatory or antiviral effects. The growing awareness and mechanistic understanding of the prothrombotic state of COVID-19 patients are driving efforts to more stringent diagnostic screening for thrombotic complications and to the early institution of antithrombotic drugs, for both the prevention and therapy of thrombotic complications. The shifting paradigm of diagnostic and treatment strategies holds significant promise to reduce the burden of thrombotic complications and ultimately improve the prognosis for patients with COVID-19.
Collapse
Affiliation(s)
- James D. McFadyen
- From the Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
- Clinical Hematology Department (J.D.M., H.S.), Alfred Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Monash University, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
| | - Hannah Stevens
- From the Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
- Clinical Hematology Department (J.D.M., H.S.), Alfred Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Monash University, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
| | - Karlheinz Peter
- Department of Cardiology (K.P.), Alfred Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Monash University, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
| |
Collapse
|
109
|
Nicolai L, Leunig A, Brambs S, Kaiser R, Weinberger T, Weigand M, Muenchhoff M, Hellmuth JC, Ledderose S, Schulz H, Scherer C, Rudelius M, Zoller M, Höchter D, Keppler O, Teupser D, Zwißler B, von Bergwelt-Baildon M, Kääb S, Massberg S, Pekayvaz K, Stark K. Immunothrombotic Dysregulation in COVID-19 Pneumonia Is Associated With Respiratory Failure and Coagulopathy. Circulation 2020; 142:1176-1189. [PMID: 32755393 PMCID: PMC7497892 DOI: 10.1161/circulationaha.120.048488] [Citation(s) in RCA: 387] [Impact Index Per Article: 96.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Supplemental Digital Content is available in the text. Background: Severe acute respiratory syndrome corona virus 2 infection causes severe pneumonia (coronavirus disease 2019 [COVID-19]), but the mechanisms of subsequent respiratory failure and complicating renal and myocardial involvement are poorly understood. In addition, a systemic prothrombotic phenotype has been reported in patients with COVID-19. Methods: A total of 62 subjects were included in our study (n=38 patients with reverse transcriptase polymerase chain reaction–confirmed COVID-19 and n=24 non–COVID-19 controls). We performed histopathologic assessment of autopsy cases, surface marker–based phenotyping of neutrophils and platelets, and functional assays for platelet, neutrophil functions, and coagulation tests, as well. Results: We provide evidence that organ involvement and prothrombotic features in COVID-19 are linked by immunothrombosis. We show that, in COVID-19, inflammatory microvascular thrombi are present in the lung, kidney, and heart, containing neutrophil extracellular traps associated with platelets and fibrin. Patients with COVID-19 also present with neutrophil-platelet aggregates and a distinct neutrophil and platelet activation pattern in blood, which changes with disease severity. Whereas cases of intermediate severity show an exhausted platelet and hyporeactive neutrophil phenotype, patients severely affected with COVID-19 are characterized by excessive platelet and neutrophil activation in comparison with healthy controls and non–COVID-19 pneumonia. Dysregulated immunothrombosis in severe acute respiratory syndrome corona virus 2 pneumonia is linked to both acute respiratory distress syndrome and systemic hypercoagulability. Conclusions: Taken together, our data point to immunothrombotic dysregulation as a key marker of disease severity in COVID-19. Further work is necessary to determine the role of immunothrombosis in COVID-19.
Collapse
Affiliation(s)
- Leo Nicolai
- Medizinische Klinik und Poliklinik I (L.N., A.L., S.B., R.K., T.W., C.S., S.K., S.M., K.P., K.S.), University Hospital Ludwig-Maximilian University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (L.N., A.L., R.K., T.W., C.S., S.K., S.M., K.P., K.S.)
| | - Alexander Leunig
- Medizinische Klinik und Poliklinik I (L.N., A.L., S.B., R.K., T.W., C.S., S.K., S.M., K.P., K.S.), University Hospital Ludwig-Maximilian University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (L.N., A.L., R.K., T.W., C.S., S.K., S.M., K.P., K.S.)
| | - Sophia Brambs
- Medizinische Klinik und Poliklinik I (L.N., A.L., S.B., R.K., T.W., C.S., S.K., S.M., K.P., K.S.), University Hospital Ludwig-Maximilian University Munich, Germany
| | - Rainer Kaiser
- Medizinische Klinik und Poliklinik I (L.N., A.L., S.B., R.K., T.W., C.S., S.K., S.M., K.P., K.S.), University Hospital Ludwig-Maximilian University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (L.N., A.L., R.K., T.W., C.S., S.K., S.M., K.P., K.S.)
| | - Tobias Weinberger
- Medizinische Klinik und Poliklinik I (L.N., A.L., S.B., R.K., T.W., C.S., S.K., S.M., K.P., K.S.), University Hospital Ludwig-Maximilian University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (L.N., A.L., R.K., T.W., C.S., S.K., S.M., K.P., K.S.)
| | - Michael Weigand
- Institute of Laboratory Medicine (M.W., D.T.), University Hospital Ludwig-Maximilian University Munich, Germany
| | - Maximilian Muenchhoff
- Virology, Max von Pettenkofer Institute (M.M., O.K.), Ludwig-Maximilian University Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich (M.M., O.K.)
| | - Johannes C Hellmuth
- Medizinische Klinik und Poliklinik III (J.C.H., M.v.B.-B.), University Hospital Ludwig-Maximilian University Munich, Germany.,German Cancer Consortium (DKTK), Munich (J.C.H., M.v.B.-B.)
| | - Stephan Ledderose
- Institute of Pathology (S.L., H.S., M.R.), Ludwig-Maximilian University Munich, Germany
| | - Heiko Schulz
- Institute of Pathology (S.L., H.S., M.R.), Ludwig-Maximilian University Munich, Germany
| | - Clemens Scherer
- Medizinische Klinik und Poliklinik I (L.N., A.L., S.B., R.K., T.W., C.S., S.K., S.M., K.P., K.S.), University Hospital Ludwig-Maximilian University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (L.N., A.L., R.K., T.W., C.S., S.K., S.M., K.P., K.S.)
| | - Martina Rudelius
- Institute of Pathology (S.L., H.S., M.R.), Ludwig-Maximilian University Munich, Germany
| | - Michael Zoller
- Department of Anesthesiology (M.Z., B.Z.), University Hospital Ludwig-Maximilian University Munich, Germany
| | | | - Oliver Keppler
- Virology, Max von Pettenkofer Institute (M.M., O.K.), Ludwig-Maximilian University Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich (M.M., O.K.)
| | - Daniel Teupser
- Institute of Laboratory Medicine (M.W., D.T.), University Hospital Ludwig-Maximilian University Munich, Germany
| | - Bernhard Zwißler
- Department of Anesthesiology (M.Z., B.Z.), University Hospital Ludwig-Maximilian University Munich, Germany
| | - Michael von Bergwelt-Baildon
- Medizinische Klinik und Poliklinik III (J.C.H., M.v.B.-B.), University Hospital Ludwig-Maximilian University Munich, Germany.,German Cancer Consortium (DKTK), Munich (J.C.H., M.v.B.-B.)
| | - Stefan Kääb
- Medizinische Klinik und Poliklinik I (L.N., A.L., S.B., R.K., T.W., C.S., S.K., S.M., K.P., K.S.), University Hospital Ludwig-Maximilian University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (L.N., A.L., R.K., T.W., C.S., S.K., S.M., K.P., K.S.)
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I (L.N., A.L., S.B., R.K., T.W., C.S., S.K., S.M., K.P., K.S.), University Hospital Ludwig-Maximilian University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (L.N., A.L., R.K., T.W., C.S., S.K., S.M., K.P., K.S.)
| | - Kami Pekayvaz
- Medizinische Klinik und Poliklinik I (L.N., A.L., S.B., R.K., T.W., C.S., S.K., S.M., K.P., K.S.), University Hospital Ludwig-Maximilian University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (L.N., A.L., R.K., T.W., C.S., S.K., S.M., K.P., K.S.)
| | - Konstantin Stark
- Medizinische Klinik und Poliklinik I (L.N., A.L., S.B., R.K., T.W., C.S., S.K., S.M., K.P., K.S.), University Hospital Ludwig-Maximilian University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Germany (L.N., A.L., R.K., T.W., C.S., S.K., S.M., K.P., K.S.)
| |
Collapse
|
110
|
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
|
111
|
Marcoux G, Laroche A, Espinoza Romero J, Boilard E. Role of platelets and megakaryocytes in adaptive immunity. Platelets 2020; 32:340-351. [PMID: 32597341 DOI: 10.1080/09537104.2020.1786043] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The immune system is comprised of two principal interconnected components called innate and adaptive immunity. While the innate immune system mounts a nonspecific response that provides protection against the spread of foreign pathogens, the adaptive immune system has developed to specifically recognize a given pathogen and lead to immunological memory. Platelets are small fragments produced from megakaryocytes in bone marrow and lungs. They circulate throughout the blood to monitor the integrity of the vasculature and to prevent bleeding. Given their large repertoire of immune receptors and inflammatory molecules, platelets and megakaryocytes can contribute to both innate and adaptive immunity. In adaptive immunity, platelets and megakaryocytes can process and present antigens to lymphocytes. Moreover, platelets, via FcγRIIA, rapidly respond to pathogens in an immune host when antibodies are present. This manuscript reviews the reported contributions of platelets and megakaryocytes with emphasis on antigen presentation and antibody response in adaptive immunity.
Collapse
Affiliation(s)
- Genevieve Marcoux
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| | - Audrée Laroche
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| | - Jenifer Espinoza Romero
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| | - Eric Boilard
- Axe Maladies Infectieuses et Inflammatoires, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada.,Département de Microbiologie-infectiologie et D'immunologie and Centre ARThrite, Université Laval, Québec, QC, Canada.,Department of Infectious Diseases and Immunity, Centre de Recherche du CHU de Québec, Québec, QC, Canada
| |
Collapse
|
112
|
Tong Q, Qiu N, Ji J, Ye L, Zhai G. Research Progress in Bioinspired Drug Delivery Systems. Expert Opin Drug Deliv 2020; 17:1269-1288. [PMID: 32543953 DOI: 10.1080/17425247.2020.1783235] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION To tackle challenges associated with traditional drug carriers, investigators have explored cells, cellular membrane, and macromolecular components including proteins and exosomes for the fabrication of delivery vehicles, owing to their excellent biocompatibility, lower toxicity, lower immunogenicity and similarities with the host. Biomacromolecule- and biomimetic nanoparticle (NP)-based drug/gene carriers are drawing immense attention, and biomimetic drug delivery systems (BDDSs) have been conceived and constructed. AREAS COVERED This review focuses on BDDS based on mammalian cells, including blood cells, cancer cells, adult stem cells, endogenous proteins, pathogens and extracellular vesicles (EVs). EXPERT OPINION Compared with traditional drug delivery systems (DDSs), BDDSs are based on biological nanocarriers, exhibiting superior biocompatibility, fewer side effects, natural targeting, and diverse modifications. In addition to directly employing natural biomaterials such as cells, proteins, pathogens and EVs as carriers, BDDSs offer these advantages by mimicking the structure of natural nanocarriers through bioengineering technologies. Furthermore, BDDSs demonstrate fewer limitations and irregularities than natural materials and can overcome several shortcomings associated with natural carriers. Although research remains ongoing to resolve these limitations, it is anticipated that BDDSs possess the potential to overcome challenges associated with traditional DDS, with a promising future in the treatment of human diseases.
Collapse
Affiliation(s)
- Qirong Tong
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University , Jinan, PR China
| | - Na Qiu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University , Jinan, PR China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University , Jinan, PR China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University , Jinan, PR China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University , Jinan, PR China
| |
Collapse
|
113
|
Zhang J, Lu Z, Xiao W, Hua T, Zheng Y, Yang M. Efficacy and Safety of Recombinant Human Thrombopoietin on Sepsis Patients With Thrombocytopenia: A Systematic Review and Meta-Analysis. Front Pharmacol 2020; 11:940. [PMID: 32714186 PMCID: PMC7344265 DOI: 10.3389/fphar.2020.00940] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/09/2020] [Indexed: 12/29/2022] Open
Abstract
Background The efficacy and safety of the administration of recombinant human thrombopoietin (rhTPO) in sepsis patients with thrombocytopenia were still inconclusive. Objectives To investigate whether rhTPO is a benefit for sepsis patients with thrombocytopenia. Methods PubMed, Cochrane library, Embase, China National Knowledge Infrastructure, and Wanfang Database were electronically searched to the randomized controlled trials (RCTs) from inception to March 4, 2020. The primary outcome was the level of platelet (PLT) on the 7th day of treatment, and secondary outcomes were 28-d mortality, the level of coagulation indicators, hepatic and renal function indicators, blood transfusion, and length of intensive care unit (ICU) stay. Results Ten RCTs involving 681 patients were included. For compared with conventional antibiotic therapy, rhTPO could significantly increase platelet counts (PCs) [standardized mean difference (SMD), 2.61; 95% confidence interval (CI), 1.28-3.94; P < 0.001], decreased 28-d mortality [relative risk (RR), 0.66; 95%CI, 0.46-0.97; P=0.03], transfusion volume of blood products and length of ICU stay. Additionally, for compared with conventional antibiotic therapy combined with intravenous immunoglobulin, the pooled results shown that rhTPO also associated with an improvement of PCs on 7th of treatment (SMD, 0.86; 95%CI, 0.54-1.17; P < 0.001), and a reduced transfusion volume of blood products. However, there were no differences in 28-d mortality and the length of ICU stay. Conclusions Current evidence shown that rhTPO could increase PCs on 7th day of treatment and reduce the transfusion volume of blood products in sepsis-related thrombocytopenia during hospitalization. The conclusions are needed to be verified indeed by more multicenter RCTs due to the limitation of the included studies.
Collapse
Affiliation(s)
- Jin Zhang
- The Second Department of Intensive Care Unit, the Second Affiliated Hospital of Anhui Medical University, Hefei, China.,The Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zongqing Lu
- The Second Department of Intensive Care Unit, the Second Affiliated Hospital of Anhui Medical University, Hefei, China.,The Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wenyan Xiao
- The Second Department of Intensive Care Unit, the Second Affiliated Hospital of Anhui Medical University, Hefei, China.,The Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tianfeng Hua
- The Second Department of Intensive Care Unit, the Second Affiliated Hospital of Anhui Medical University, Hefei, China.,The Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yao Zheng
- The Second Department of Intensive Care Unit, the Second Affiliated Hospital of Anhui Medical University, Hefei, China.,The Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Min Yang
- The Second Department of Intensive Care Unit, the Second Affiliated Hospital of Anhui Medical University, Hefei, China.,The Laboratory of Cardiopulmonary Resuscitation and Critical Care Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
114
|
Cheng J, Cao X, Hanif Q, Pi L, Hu L, Huang Y, Lan X, Lei C, Chen H. Integrating Genome-Wide CNVs Into QTLs and High Confidence GWAScore Regions Identified Positional Candidates for Sheep Economic Traits. Front Genet 2020; 11:569. [PMID: 32655616 PMCID: PMC7325882 DOI: 10.3389/fgene.2020.00569] [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: 01/16/2020] [Accepted: 05/11/2020] [Indexed: 12/21/2022] Open
Abstract
Copy number variations (CNVs) are important source of genetic variation, which can affect diverse economic traits through a variety of mechanisms. In addition, genome scan can identify many quantitative trait loci (QTLs) for the economic traits, while genome-wide association studies (GWAS) can localize genetic variants associated with the phenotypic variations. Here, we developed a method called GWAScore which collected GWAS summary data to identify potential candidates, and integrated CNVs into QTLs and high confidence GWAScore regions to detect crucial CNV markers for sheep growth traits. We got 197 candidate genes which were overlapping with the candidate CNVs. Some crucial genes (MYLK3, TTC29, HERC6, ABCG2, RUNX1, etc.) showed significantly elevated GWAScore peaks than other candidate genes. In this study, we developed the GWAScore method to excavate the potential value of candidate genes as markers for the sheep molecular breeding.
Collapse
Affiliation(s)
- Jie Cheng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xiukai Cao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Quratulain Hanif
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China.,Computational Biology Lab, National Institute for Biotechnology and Genetic Engineering, Faisalabad, Pakistan.,Pakistan Institute of Engineering and Applied Sciences, Islamabad, Pakistan
| | - Li Pi
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
| | - Linyong Hu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
| | - Yongzhen Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xianyong Lan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Hong Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| |
Collapse
|
115
|
Marrero MG, Field SL, Skibiel AL, Dado-Senn B, Driver JP, Laporta J. Increasing serotonin bioavailability alters gene expression in peripheral leukocytes and lymphoid tissues of dairy calves. Sci Rep 2020; 10:9712. [PMID: 32546841 PMCID: PMC7297988 DOI: 10.1038/s41598-020-66326-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 05/08/2020] [Indexed: 12/26/2022] Open
Abstract
Dairy calves are born with a naïve immune system, making the pre-weaning phase a critical window for immune development. In the U.S., 40-60% of dairy farms feed milk replacer to pre-weaned calves, which are devoid of bioactive factors with immunological roles. Serotonin is a bioactive factor with immunoregulatory properties naturally produced by the calf and present in milk. Human and rodent immune cells express the serotonin machinery, but little is known about the role of serotonin in the bovine immune system. Supplementing milk replacer with 5-hydroxytryptophan (serotonin precursor) or fluoxetine (reuptake inhibitor) increases serotonin bioavailability. We hypothesized that increased serotonin bioavailability promotes serotonergic signaling and modulates the expression of immune related genes in peripheral leukocytes and immune-related tissues of dairy calves. The present experiment targeted candidate genes involved in serotonin production, metabolism, transport, signaling and immune regulation. We established that bovine peripheral leukocytes express all known serotonin receptors, and can synthesize, uptake and degrade serotonin due to the expression of serotonin metabolism-related genes. Indeed, we showed that increasing serotonin bioavailability alters gene expression of serotonin receptors and immune-related genes. Further research will determine whether manipulation of the serotonin pathway could be a feasible approach to bolster dairy calves' immune system.
Collapse
Affiliation(s)
- M G Marrero
- Department of Animal Sciences, University of Florida, Florida, USA
| | - S L Field
- Department of Animal Sciences, University of Florida, Florida, USA
| | - A L Skibiel
- Department of Animal Sciences, University of Florida, Florida, USA
- Department of Animal and Veterinary Science, University of Idaho, Idaho, USA
| | - B Dado-Senn
- Department of Animal Sciences, University of Florida, Florida, USA
| | - J P Driver
- Department of Animal Sciences, University of Florida, Florida, USA
| | - J Laporta
- Department of Animal Sciences, University of Florida, Florida, USA.
| |
Collapse
|
116
|
Charlton JR, Baldelomar EJ, deRonde KA, Cathro HP, Charlton NP, Criswell SJ, Hyatt DM, Nam S, Pearl V, Bennett KM. Nephron loss detected by MRI following neonatal acute kidney injury in rabbits. Pediatr Res 2020; 87:1185-1192. [PMID: 31805577 PMCID: PMC7255918 DOI: 10.1038/s41390-019-0684-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 10/14/2019] [Accepted: 10/18/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Acute kidney injury affects nearly 30% of preterm neonates in the intensive care unit. We aimed to determine whether nephrotoxin-induced AKI disrupted renal development assessed by imaging (CFE-MRI). METHODS Neonatal New Zealand rabbits received indomethacin and gentamicin (AKI) or saline (control) for four days followed by cationic ferritin (CF) at six weeks. Ex vivo images were acquired using a gradient echo pulse sequence on 7 T MRI. Glomerular number (Nglom) and apparent glomerular volume (aVglom) were determined. CF toxicity was assessed at two and 28 days in healthy rabbits. RESULTS Nglom was lower in the AKI group as compared to controls (74,034 vs 198,722, p < 0.01). aVglom was not different (AKI: 7.3 × 10-4 vs control: 6.2 × 10-4 mm3, p = 0.69). AKI kidneys had a band of glomeruli distributed radially in the cortex that were undetectable by MRI. Following CF injection, there was no difference in body or organ weights except for the liver, and transient changes in serum iron, platelets and white blood cell count. CONCLUSIONS Brief nephrotoxin exposure during nephrogenesis results in fewer glomeruli and glomerular maldevelopment in a unique pattern detectable by MRI. Whole kidney evaluation by CFE-MRI may provide an important tool to understand the development of CKD following AKI.
Collapse
Affiliation(s)
- Jennifer R Charlton
- Department of Pediatrics, Division of Nephrology, University of Virginia, Charlottesville, VA, USA.
| | | | - Kimberly A deRonde
- Department of Pediatrics, Division of Nephrology, University of Virginia, Charlottesville, VA, USA
| | - Helen P Cathro
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Nathan P Charlton
- Department of Emergency Medicine, Division of Medical Toxicology, University of Virginia, Charlottesville, VA, USA
| | - Stacey J Criswell
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA
| | - Dylan M Hyatt
- School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Sejin Nam
- Department of Physics, University of Hawaii, Honolulu, HI, USA
| | - Valeria Pearl
- Department of Pediatrics, Division of Nephrology, University of Virginia, Charlottesville, VA, USA
| | | |
Collapse
|
117
|
Leiter O, Walker TL. Platelets in Neurodegenerative Conditions-Friend or Foe? Front Immunol 2020; 11:747. [PMID: 32431701 PMCID: PMC7214916 DOI: 10.3389/fimmu.2020.00747] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
It is now apparent that platelet function is more diverse than originally thought, shifting the view of platelets from blood cells involved in hemostasis and wound healing to major contributors to numerous regulatory processes across different tissues. Given their intriguing ability to store, produce and release distinct subsets of bioactive molecules, including intercellular signaling molecules and neurotransmitters, platelets may play an important role in orchestrating healthy brain function. Conversely, a number of neurodegenerative conditions have recently been associated with platelet dysfunction, further highlighting the tissue-independent role of these cells. In this review we summarize the requirements for platelet-neural cell communication with a focus on neurodegenerative diseases, and discuss the therapeutic potential of healthy platelets and the proteins which they release to counteract these conditions.
Collapse
Affiliation(s)
- Odette Leiter
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Tara L Walker
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
118
|
Tessandier N, Melki I, Cloutier N, Allaeys I, Miszta A, Tan S, Milasan A, Michel S, Benmoussa A, Lévesque T, Côté F, McKenzie SE, Gilbert C, Provost P, Brisson AR, Wolberg AS, Fortin PR, Martel C, Boilard É. Platelets Disseminate Extracellular Vesicles in Lymph in Rheumatoid Arthritis. Arterioscler Thromb Vasc Biol 2020; 40:929-942. [PMID: 32102567 PMCID: PMC8073225 DOI: 10.1161/atvbaha.119.313698] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The lymphatic system is a circulatory system that unidirectionally drains the interstitial tissue fluid back to blood circulation. Although lymph is utilized by leukocytes for immune surveillance, it remains inaccessible to platelets and erythrocytes. Activated cells release submicron extracellular vesicles (EV) that transport molecules from the donor cell. In rheumatoid arthritis, EV accumulate in the joint where they can interact with numerous cellular lineages. However, whether EV can exit the inflamed tissue to recirculate is unknown. Here, we investigated whether vascular leakage that occurs during inflammation could favor EV access to the lymphatic system. Approach and Results: Using an in vivo model of autoimmune inflammatory arthritis, we show that there is an influx of platelet EV, but not EV from erythrocytes or leukocytes, in joint-draining lymph. In contrast to blood platelet EV, lymph platelet EV lacked mitochondrial organelles and failed to promote coagulation. Platelet EV influx in lymph was consistent with joint vascular leakage and implicated the fibrinogen receptor α2bβ3 and platelet-derived serotonin. CONCLUSIONS These findings show that platelets can disseminate their EV in fluid that is inaccessible to platelets and beyond the joint in this disease.
Collapse
Affiliation(s)
- Nicolas Tessandier
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
| | - Imene Melki
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
| | - Nathalie Cloutier
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
| | - Isabelle Allaeys
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
| | - Adam Miszta
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill (A.M., A.S.W.)
- Montreal Heart Institute, Quebec, Canada (A.M., C.M.)
| | - Sisareuth Tan
- Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, Pessac, France (S.T., A.R.B.)
| | - Andreea Milasan
- Department of Medicine, Faculty of Medicine (A.M., C.M.), Université de Montréal, Quebec, Canada
| | - Sara Michel
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
| | - Abderrahim Benmoussa
- Department of Nutrition, CHU Sainte-Justine (A.B.), Université de Montréal, Quebec, Canada
| | - Tania Lévesque
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
| | - Francine Côté
- Institut Imagine, Inserm U1163, Laboratoire Olivier Hermine, Paris, France (F.C.)
| | - Steven E McKenzie
- Cardeza Foundation for Hematological Research, Thomas Jefferson University, Philadelphia, PA (S.E.M.)
| | - Caroline Gilbert
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
| | - Patrick Provost
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
| | - Alain R Brisson
- Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, Pessac, France (S.T., A.R.B.)
| | - Alisa S Wolberg
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill (A.M., A.S.W.)
| | - Paul R Fortin
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Axe maladies infectieuses et inflammatoires, Centre de recherche du CHU de Québec-Université Laval, Québec, Canada (P.R.F., E.B.)
| | - Catherine Martel
- Department of Medicine, Faculty of Medicine (A.M., C.M.), Université de Montréal, Quebec, Canada
- Montreal Heart Institute, Quebec, Canada (A.M., C.M.)
| | - Éric Boilard
- From the Centre de recherche du CHU de Québec, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, QC, Canada (N.T., I.M., N.C., I.A., S.M., T.L., C.G., P.P., P.R.F., E.B.)
- Axe maladies infectieuses et inflammatoires, Centre de recherche du CHU de Québec-Université Laval, Québec, Canada (P.R.F., E.B.)
| |
Collapse
|
119
|
Platelets: Mechanistic and Diagnostic Significance in Transplantation. CURRENT TRANSPLANTATION REPORTS 2020. [DOI: 10.1007/s40472-020-00272-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Abstract
Purpose of Review
In addition to their function in coagulation, platelets recently have been recognized as an important component of innate immune responses. This review relates salient immune functions of platelets to transplants.
Recent Findings
Platelets are critical bridges between vascular endothelium and leukocytes. Real-time imaging of platelets has demonstrated that platelets rapidly adhere to vascular endothelium and form a nidus for attachment of neutrophils and then monocytes. However, the majority of platelets subsequently release from endothelium and return to the circulation in an activated state. These recycled platelets have the potential to transport proteins and RNA from the graft to the recipient. Some of the platelets that return to the circulation are attached to leukocytes.
Summary
Platelets have the potential to modulate many elements of the graft and the immune response from the time of organ retrieval through ischemia-reperfusion to acute and chronic rejection. Beyond mechanistic considerations, assays that detect changes in platelet protein or RNA expression could be used to monitor early inflammatory responses in transplants.
Collapse
|
120
|
Kniewallner KM, de Sousa DMB, Unger MS, Mrowetz H, Aigner L. Platelets in Amyloidogenic Mice Are Activated and Invade the Brain. Front Neurosci 2020; 14:129. [PMID: 32194368 PMCID: PMC7063083 DOI: 10.3389/fnins.2020.00129] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/31/2020] [Indexed: 12/20/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease with a complex and not fully understood pathogenesis. Besides brain-intrinsic hallmarks such as abnormal deposition of harmful proteins, i.e., amyloid beta in plaques and hyperphosphorylated Tau in neurofibrillary tangles, blood-derived elements, in particular, platelets have been discussed to be involved in AD pathogenesis. The underlying mechanisms, however, are rather unexplored. Here, we investigate a potential role of platelets in an AD transgenic animal model with severe amyloid plaque formation, the APP-PS1 transgenic mice, and analyzed the presence, spatial location and activation status of platelets within the brain. In APP-PS1 mice, a higher number of platelets were located within the brain parenchyma, i.e., outside the cerebral blood vessels compared to WT controls. Such platelets were activated according to the expression of the platelet activation marker CD62P and to morphological hallmarks such as membrane protrusions. In the brain, platelets were in close contact exclusively with astrocytes suggesting an interaction between these two cell types. In the bloodstream, although the percentage of activated platelets did not differ between transgenic and age-matched control animals, APP-PS1 blood-derived platelets showed remarkable ultrastructural peculiarities in platelet-specific organelles such as the open canalicular system (OCS). This work urges for further investigations on platelets and their yet unknown functional roles in the brain, which might go beyond AD pathogenesis and be relevant for various age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Kathrin M Kniewallner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Diana M Bessa de Sousa
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Michael S Unger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Heike Mrowetz
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
121
|
Kopeikina E, Dukhinova M, Yung AWY, Veremeyko T, Kuznetsova IS, Lau TYB, Levchuk K, Ponomarev ED. Platelets promote epileptic seizures by modulating brain serotonin level, enhancing neuronal electric activity, and contributing to neuroinflammation and oxidative stress. Prog Neurobiol 2020; 188:101783. [PMID: 32142857 DOI: 10.1016/j.pneurobio.2020.101783] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 02/04/2020] [Accepted: 02/24/2020] [Indexed: 11/28/2022]
Abstract
The drugs currently available for treating epilepsy are only partially effective in managing this condition. Therefore, it is crucial to investigate new pathways that induce and promote epilepsy development. Previously, we found that platelets interact with neuronal glycolipids and actively secrete pro-inflammatory mediators during central nervous system (CNS) pathological conditions such as neuroinflammation and traumatic brain injury (TBI). These factors increase the permeability of the blood-brain barrier (BBB), which may create a predisposition to epileptic seizures. In this study, we demonstrated that platelets substantially enhanced epileptic seizures in a mouse model of pentylenetetrazole (PTZ) -induced seizures. We found that platelets actively secreted serotonin, contributed to increased BBB permeability, and were present in the CNS parenchyma during epileptic seizures. Furthermore, platelets directly stimulated neuronal electric activity and induced the expression of specific genes related to early neuronal response, neuroinflammation, and oxidative phosphorylation, leading to oxidative stress in neurons. The intracranial injection of physiological numbers of platelets that mimicked TBI-associated bleeding was sufficient to induce severe seizures, which resembled conventional PTZ-induced epileptic activity. These findings highlight a conceptually new role of platelets in the development of epileptic seizures, and indicate a potential new therapeutic approach targeting platelets to prevent and treat epilepsy.
Collapse
Affiliation(s)
- Ekaterina Kopeikina
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Marina Dukhinova
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Amanda W Y Yung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Tatyana Veremeyko
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Inna S Kuznetsova
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Thomas Y B Lau
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Kseniia Levchuk
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Eugene D Ponomarev
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong.
| |
Collapse
|
122
|
Karhausen J, Choi HW, Maddipati KR, Mathew JP, Ma Q, Boulaftali Y, Lee RH, Bergmeier W, Abraham SN. Platelets trigger perivascular mast cell degranulation to cause inflammatory responses and tissue injury. SCIENCE ADVANCES 2020; 6:eaay6314. [PMID: 32206714 PMCID: PMC7080499 DOI: 10.1126/sciadv.aay6314] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 12/20/2019] [Indexed: 06/08/2023]
Abstract
Platelet responses have been associated with end-organ injury and mortality following complex insults such as cardiac surgery, but how platelets contribute to these pathologies remains unclear. Our studies originated from the observation of microvascular platelet retention in a rat cardiac surgery model. Ensuing work supported the proximity of platelet aggregates with perivascular mast cells (MCs) and demonstrated that platelet activation triggered systemic MC activation. We then identified platelet activating factor (PAF) as the platelet-derived mediator stimulating MCs and, using chimeric animals with platelets defective in PAF generation or MCs lacking PAF receptor, defined the role of this platelet-MC interaction for vascular leakage, shock, and tissue inflammation. In application of these findings, we demonstrated that inhibition of platelet activation in modeled cardiac surgery blunted MC-dependent inflammation and tissue injury. Together, our work identifies a previously undefined mechanism of inflammatory augmentation, in which platelets trigger local and systemic responses through activation of perivascular MCs.
Collapse
Affiliation(s)
- Jörn Karhausen
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Hae Woong Choi
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
- Department of Life Sciences, Korea University, Seoul 02841, South Korea
| | | | - Joseph P. Mathew
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Qing Ma
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Yacine Boulaftali
- Université Paris Diderot, Sorbonne Paris Cité, Laboratory of Vascular Translational Science, U1148 Institute National de la Santé et de la Recherche Medicale (INSERM), Paris, France
| | - Robert Hugh Lee
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, USA
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, USA
- UNC Center for Blood Research, University of North Carolina, Chapel Hill, NC, USA
| | - Soman N. Abraham
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
- Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore, Singapore
| |
Collapse
|
123
|
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease in which a variety of circulating pro-inflammatory cells and dysregulated molecules are involved in disease aetiology and progression. Platelets are an important cellular element in the circulation that can bind several dysregulated molecules (such as collagen, thrombin and fibrinogen) that are present both in the synovium and the circulation of patients with RA. Platelets not only respond to dysregulated molecules in their environment but also transport and express their own inflammatory mediators, and serve as regulators at the boundary between haemostasis and immunity. Activated platelets also produce microparticles, which further convey signalling molecules and receptors to the synovium and circulation, thereby positioning these platelet-derived particles as strategic regulators of inflammation. These diverse functions come together to make platelets facilitators of cellular crosstalk in RA. Thus, the receptor functions, ligand binding potential and dysregulated signalling pathways in platelets are becoming increasingly important for treatment in RA. This Review aims to highlight the role of platelets in RA and the need to closely examine platelets as health indicators when designing effective pharmaceutical targets in this disease.
Collapse
|
124
|
Rayes J, Bourne JH, Brill A, Watson SP. The dual role of platelet-innate immune cell interactions in thrombo-inflammation. Res Pract Thromb Haemost 2020; 4:23-35. [PMID: 31989082 PMCID: PMC6971330 DOI: 10.1002/rth2.12266] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/04/2019] [Accepted: 09/09/2019] [Indexed: 12/11/2022] Open
Abstract
Beyond their role in hemostasis and thrombosis, platelets are increasingly recognized as key regulators of the inflammatory response under sterile and infectious conditions. Both platelet receptors and secretion are critical for these functions and contribute to their interaction with the endothelium and innate immune system. Platelet-leukocyte interactions are increased in thrombo-inflammatory diseases and are sensitive biomarkers for platelet activation and targets for the development of new therapies. The crosstalk between platelets and innate immune cells promotes thrombosis, inflammation, and tissue damage. However, recent studies have shown that these interactions also regulate the resolution of inflammation, tissue repair, and wound healing. Many of the platelet and leukocyte receptors involved in these bidirectional interactions are not selective for a subset of immune cells. However, specific heterotypic interactions occur in different vascular beds and inflammatory conditions, raising the possibility of disease- and organ-specific pathways of intervention. In this review, we highlight and discuss prominent and emerging interrelationships between platelets and innate immune cells and their dual role in the regulation of the inflammatory response in sterile and infectious thrombo-inflammatory diseases. A better understanding of the functional relevance of these interactions in different vascular beds may provide opportunities for successful therapeutic interventions to regulate the development, progression, and chronicity of various pathological processes.
Collapse
Affiliation(s)
- Julie Rayes
- Institute of Cardiovascular SciencesCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)Universities of Birmingham and NottinghamThe MidlandsUK
| | - Joshua H. Bourne
- Institute of Cardiovascular SciencesCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Alexander Brill
- Institute of Cardiovascular SciencesCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)Universities of Birmingham and NottinghamThe MidlandsUK
- Department of PathophysiologySechenov First Moscow State Medical University (Sechenov University)MoscowRussia
| | - Steve P. Watson
- Institute of Cardiovascular SciencesCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)Universities of Birmingham and NottinghamThe MidlandsUK
| |
Collapse
|
125
|
|
126
|
Distler JHW, Györfi AH, Ramanujam M, Whitfield ML, Königshoff M, Lafyatis R. Shared and distinct mechanisms of fibrosis. Nat Rev Rheumatol 2019; 15:705-730. [DOI: 10.1038/s41584-019-0322-7] [Citation(s) in RCA: 197] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
|
127
|
Washington AV, Esponda O, Gibson A. Platelet biology of the rapidly failing lung. Br J Haematol 2019; 188:641-651. [PMID: 31696941 DOI: 10.1111/bjh.16315] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/23/2019] [Accepted: 08/24/2019] [Indexed: 12/15/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by a rapid-onset respiratory failure with a mortality rate of approximately 40%. This physiologic inflammatory process is mediated by disruption of the alveolar-vascular interface, leading to pulmonary oedema and impaired oxygen exchange, which often warrants mechanical ventilation to increase survival in the acute setting. One of the least understood aspects of ARDS is the role of the platelets in this process. Platelets, which protect vascular integrity, play a pivotal role in the progression and resolution of ARDS. The recent substantiation of the age-old theory that megakaryocytes are found in the lungs has rejuvenated interest in and raised new questions about the importance of platelets for pulmonary function. In addition to primary haemostasis, platelets provide a myriad of inflammatory functions that are poised to aid the innate immune system. This review focuses on the evidence for regulatory roles of platelets in pulmonary inflammation, with an emphasis on two receptors, CLEC-2 and TLT-1. Studies of these receptors identify novel pathways through which platelets may regulate vascular integrity and inflammation in the lungs, thereby influencing the development of ARDS.
Collapse
Affiliation(s)
- A Valance Washington
- Department of Biology, Molecular Science Research Center, University of Puerto Rico-Rio Piedras, San Juan, PR, USA
| | | | | |
Collapse
|
128
|
Margraf A, Zarbock A. Platelets in Inflammation and Resolution. THE JOURNAL OF IMMUNOLOGY 2019; 203:2357-2367. [DOI: 10.4049/jimmunol.1900899] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 08/30/2019] [Indexed: 12/22/2022]
|
129
|
Abstract
The rate-limiting enzyme in serotonin synthesis is tryptophan hydroxylase (TPH). There are two independent serotonin systems in the body characterized by two isoforms of TPH, TPH1 and TPH2. While TPH2 synthesizes serotonin in the brain, TPH1 is expressed in the gut and in other peripheral tissues and supplies platelets in the circulation with serotonin. This duality of the serotonin system is enforced by the blood-brain barrier which is impermeable for serotonin. In the brain serotonin acts as neurotransmitter and is a main target for the treatment of psychiatric disorders. In the periphery it is released by platelets at the site of activation and elicits numerous physiological effects. TPH1 deficient mice were shown to be protected from diverse diseases including hemostatic, inflammatory, fibrotic, gastrointestinal, and metabolic disorders and therefore serotonin synthesis inhibition emerged as a reasonable therapeutic paradigm. Recently the first TPH inhibitor, telotristat ethyl, came on the market for the treatment of carcinoid syndrome. This review summarizes the state of development and the therapeutic opportunities of such compounds.
Collapse
Affiliation(s)
- Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125 Berlin-Buch, Germany; University of Lübeck, Institute for Biology, Ratzeburger Allee 160, 23562 Lübeck, Germany; Charité University Medicine, Charitéplatz 1, 10117 Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site, Berlin, Germany.
| |
Collapse
|
130
|
Affiliation(s)
- Dermot Cox
- Molecular & Cellular Therapeutics Royal College of Surgeons in Ireland Dublin Ireland
| |
Collapse
|
131
|
Guo L, Rondina MT. The Era of Thromboinflammation: Platelets Are Dynamic Sensors and Effector Cells During Infectious Diseases. Front Immunol 2019; 10:2204. [PMID: 31572400 PMCID: PMC6753373 DOI: 10.3389/fimmu.2019.02204] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/30/2019] [Indexed: 12/12/2022] Open
Abstract
Platelets are anucleate cells produced by megakaryocytes. In recent years, a robust body of literature supports the evolving role of platelets as key sentinel and effector cells in infectious diseases, especially critical in bridging hemostatic, inflammatory, and immune continuums. Upon intravascular pathogen invasion, platelets can directly sense viral, parasitic, and bacterial infections through pattern recognition receptors and integrin receptors or pathogen: immunoglobulin complexes through Fc and complement receptors—although our understanding of these interactions remains incomplete. Constantly scanning for areas of injury or inflammation as they circulate in the vasculature, platelets also indirectly respond to pathogen invasion through interactions with leukocytes and the endothelium. Following antigen recognition, platelets often become activated. Through a diverse repertoire of mechanisms, activated platelets can directly sequester or kill pathogens, or facilitate pathogen clearance by activating macrophages and neutrophils, promoting neutrophil extracellular traps (NETs) formation, forming platelet aggregates and microthrombi. At times, however, platelet activation may also be injurious to the host, exacerbating inflammation and promoting endothelial damage and thrombosis. There are many gaps in our understandings of the role of platelets in infectious diseases. However, with the emergence of advanced technologies, our knowledge is increasing. In the current review, we mainly discuss these evolving roles of platelets under four different infectious pathogen infections, of which are dengue, malaria, Esterichia coli (E. coli) and staphylococcus aureus S. aureus, highlighting the complex interplay of these processes with hemostatic and thrombotic pathways.
Collapse
Affiliation(s)
- Li Guo
- University of Utah Molecular Medicine Program, Salt Lake City, UT, United States
| | - Matthew T Rondina
- University of Utah Molecular Medicine Program, Salt Lake City, UT, United States.,Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States.,Department of Pathology, University of Utah, Salt Lake City, UT, United States.,George E. Wahlen VAMC Department of Internal Medicine and GRECC, Salt Lake City, UT, United States
| |
Collapse
|
132
|
Abstract
Dysregulation of lymphocyte function, accumulation of autoantibodies and defective clearance of circulating immune complexes and apoptotic cells are hallmarks of systemic lupus erythematosus (SLE). Moreover, it is now evident that an intricate interplay between the adaptive and innate immune systems contributes to the pathogenesis of SLE, ultimately resulting in chronic inflammation and organ damage. Platelets circulate in the blood and are chiefly recognized for their role in the prevention of bleeding and promotion of haemostasis; however, accumulating evidence points to a role for platelets in both adaptive and innate immunity. Through a broad repertoire of receptors, platelets respond promptly to immune complexes, complement and damage-associated molecular patterns, and represent a major reservoir of immunomodulatory molecules in the circulation. Furthermore, evidence suggests that platelets are activated in patients with SLE, and that they could contribute to the circulatory autoantigenic load through the release of microparticles and mitochondrial antigens. Herein, we highlight how platelets contribute to the immune response and review evidence implicating platelets in the pathogenesis of SLE.
Collapse
|
133
|
Kerrigan SW, Devine T, Fitzpatrick G, Thachil J, Cox D. Early Host Interactions That Drive the Dysregulated Response in Sepsis. Front Immunol 2019; 10:1748. [PMID: 31447831 PMCID: PMC6691039 DOI: 10.3389/fimmu.2019.01748] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/10/2019] [Indexed: 01/18/2023] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. While many individual cells and systems in the body are involved in driving the excessive and sometimes sustained host response, pathogen engagement with endothelial cells and platelets early in sepsis progression, are believed to be key. Significant progress has been made in establishing key molecular interactions between platelets and pathogens and endothelial cells and pathogens. This review will explore the growing number of compensatory connections between bacteria and viruses with platelets and endothelial cells and how a better understanding of these interactions are informing the field of potential novel ways to treat the dysregulated host response during sepsis.
Collapse
Affiliation(s)
- Steven W Kerrigan
- Cardiovascular Infection Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland.,Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tatyana Devine
- Cardiovascular Infection Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Glenn Fitzpatrick
- Cardiovascular Infection Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jecko Thachil
- Department of Haematology, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Dermot Cox
- Cardiovascular Infection Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
134
|
Orsini F, Fumagalli S, Császár E, Tóth K, De Blasio D, Zangari R, Lénárt N, Dénes Á, De Simoni MG. Mannose-Binding Lectin Drives Platelet Inflammatory Phenotype and Vascular Damage After Cerebral Ischemia in Mice via IL (Interleukin)-1α. Arterioscler Thromb Vasc Biol 2019; 38:2678-2690. [PMID: 30354247 PMCID: PMC6221395 DOI: 10.1161/atvbaha.118.311058] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Circulating complement factors are activated by tissue damage and contribute to acute brain injury. The deposition of MBL (mannose-binding lectin), one of the initiators of the lectin complement pathway, on the cerebral endothelium activated by ischemia is a major pathogenic event leading to brain injury. The molecular mechanisms through which MBL influences outcome after ischemia are not understood yet. Approach and Results— Here we show that MBL-deficient (MBL−/−) mice subjected to cerebral ischemia display better flow recovery and less plasma extravasation in the brain than wild-type mice, as assessed by in vivo 2-photon microscopy. This results in reduced vascular dysfunction as shown by the shift from a pro- to an anti-inflammatory vascular phenotype associated with MBL deficiency. We also show that platelets directly bind MBL and that platelets from MBL−/− mice have reduced inflammatory phenotype as indicated by reduced IL-1α (interleukin-1α) content, as early as 6 hours after ischemia. Cultured human brain endothelial cells subjected to oxygen-glucose deprivation and exposed to platelets from MBL−/− mice present less cell death and lower CXCL1 (chemokine [C-X-C motif] ligand 1) release (downstream to IL-1α) than those exposed to wild-type platelets. In turn, MBL deposition on ischemic vessels significantly decreases after ischemia in mice treated with IL-1 receptor antagonist compared with controls, indicating a reciprocal interplay between MBL and IL-1α facilitating endothelial damage. Conclusions— We propose MBL as a hub of pathogenic vascular events. It acts as an early trigger of platelet IL-1α release, which in turn favors MBL deposition on ischemic vessels promoting an endothelial pro-inflammatory phenotype.
Collapse
Affiliation(s)
- Franca Orsini
- From the Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy (F.O., S.F., D.D.B., R.Z., M.-G.D.S.)
| | - Stefano Fumagalli
- From the Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy (F.O., S.F., D.D.B., R.Z., M.-G.D.S.)
| | - Eszter Császár
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary (E.C., K.T., N.L., A.D.)
| | - Krisztina Tóth
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary (E.C., K.T., N.L., A.D.)
| | - Daiana De Blasio
- From the Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy (F.O., S.F., D.D.B., R.Z., M.-G.D.S.)
| | - Rosalia Zangari
- From the Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy (F.O., S.F., D.D.B., R.Z., M.-G.D.S.)
| | - Nikolett Lénárt
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary (E.C., K.T., N.L., A.D.)
| | - Ádám Dénes
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary (E.C., K.T., N.L., A.D.)
| | - Maria-Grazia De Simoni
- From the Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy (F.O., S.F., D.D.B., R.Z., M.-G.D.S.)
| |
Collapse
|
135
|
Mezger M, Nording H, Sauter R, Graf T, Heim C, von Bubnoff N, Ensminger SM, Langer HF. Platelets and Immune Responses During Thromboinflammation. Front Immunol 2019; 10:1731. [PMID: 31402914 PMCID: PMC6676797 DOI: 10.3389/fimmu.2019.01731] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/09/2019] [Indexed: 12/17/2022] Open
Abstract
Besides mediating hemostatic functions, platelets are increasingly recognized as important players of inflammation. Data from experiments in mice and men revealed various intersection points between thrombosis, hemostasis, and inflammation, which are addressed and discussed in this review in detail. One such example is the intrinsic coagulation cascade that is initiated after platelet activation thereby further propagating and re-enforcing wound healing or thrombus formation but also contributing to the pathophysiology of severe diseases. FXII of the intrinsic pathway connects platelet activation with the coagulation cascade during immune reactions. It can activate the contact system thereby either creating an inflammatory state or accelerating inflammation. Recent insights into platelet biology could show that platelets are equipped with complement receptors. Platelets are important for tissue remodeling after injury has been inflicted to the endothelial barrier and to the subendothelial tissue. Thus, platelets are increasingly recognized as more than just cells relevant for bleeding arrest. Future insights into platelet biology are to be expected. This research will potentially offer novel opportunities for therapeutic intervention in diseases featuring platelet abundance.
Collapse
Affiliation(s)
- Matthias Mezger
- University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Henry Nording
- University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Reinhard Sauter
- University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Tobias Graf
- University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Christian Heim
- Department of Cardiac Surgery, University Hospital Erlangen, Erlangen, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, Medical Center, University of Schleswig-Holstein, Lübeck, Germany
| | - Stephan M Ensminger
- Department of Cardiac and Thoracic Vascular Surgery, University Heart Center Lübeck, Lübeck, Germany
| | - Harald F Langer
- University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany
| |
Collapse
|
136
|
Assinger A, Schrottmaier WC, Salzmann M, Rayes J. Platelets in Sepsis: An Update on Experimental Models and Clinical Data. Front Immunol 2019; 10:1687. [PMID: 31379873 PMCID: PMC6650595 DOI: 10.3389/fimmu.2019.01687] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/04/2019] [Indexed: 12/22/2022] Open
Abstract
Beyond their important role in hemostasis, platelets play a crucial role in inflammatory diseases. This becomes apparent during sepsis, where platelet count and activation correlate with disease outcome and survival. Sepsis is caused by a dysregulated host response to infection, leading to organ dysfunction, permanent disabilities, or death. During sepsis, tissue injury results from the concomitant uncontrolled activation of the complement, coagulation, and inflammatory systems as well as platelet dysfunction. The balance between the systemic inflammatory response syndrome (SIRS) and the compensatory anti-inflammatory response (CARS) regulates sepsis outcome. Persistent thrombocytopenia is considered as an independent risk factor of mortality in sepsis, although it is still unclear whether the drop in platelet count is the cause or the consequence of sepsis severity. The role of platelets in sepsis development and progression was addressed in different experimental in vivo models, particularly in mice, that represent various aspects of human sepsis. The immunomodulatory function of platelets depends on the experimental model, time, and type of infection. Understanding the molecular mechanism of platelet regulation in inflammation could bring us one step closer to understand this important aspect of primary hemostasis which drives thrombotic as well as bleeding complications in patients with sterile and infectious inflammation. In this review, we summarize the current understanding of the contribution of platelets to sepsis severity and outcome. We highlight the differences between platelet receptors in mice and humans and discuss the potential and limitations of animal models to study platelet-related functions in sepsis.
Collapse
Affiliation(s)
- Alice Assinger
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | - Manuel Salzmann
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Julie Rayes
- Institute of Cardiovascular Science, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
137
|
Liu X, Gorzelanny C, Schneider SW. Platelets in Skin Autoimmune Diseases. Front Immunol 2019; 10:1453. [PMID: 31333641 PMCID: PMC6620619 DOI: 10.3389/fimmu.2019.01453] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 06/10/2019] [Indexed: 12/11/2022] Open
Abstract
Systemic lupus erythematosus (SLE), systemic sclerosis (SSc), and small vessel vasculitis are three autoimmune diseases frequently manifested in the skin. They share common pathogenic features, including production of autoantibodies, loss of tolerance to self-antigens, tissue necrosis and fibrosis, vasculopathy and activation of the coagulation system. Platelets occupy a central part within the coagulation cascade and are well-recognized for their hemostatic role. However, recent cumulative evidence implicates their additional and multifaceted immunoregulatory functions. Platelets express immune receptors and they store growth factors, cytokines, and chemokines in their granules enabling a significant contribution to inflammation. A plethora of activating triggers such as damage associated molecular patterns (DAMPs) released from damaged endothelial cells, immune complexes, or complement effector molecules can mediate platelet activation. Activated platelets further foster an inflammatory environment and the crosstalk with the endothelium and leukocytes by the release of immunoactive molecules and microparticles. Further insight into the pathogenic implications of platelet activation will pave the way for new therapeutic strategies targeting autoimmune diseases. In this review, we discuss the inflammatory functions of platelets and their mechanistic contribution to the pathophysiology of SSc, ANCA associated small vessel vasculitis and other autoimmune diseases affecting the skin.
Collapse
Affiliation(s)
- Xiaobo Liu
- Department of Dermatology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Gorzelanny
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan W Schneider
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
138
|
Menikou S, Langford PR, Levin M. Kawasaki Disease: The Role of Immune Complexes Revisited. Front Immunol 2019; 10:1156. [PMID: 31263461 PMCID: PMC6584825 DOI: 10.3389/fimmu.2019.01156] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 05/07/2019] [Indexed: 01/09/2023] Open
Abstract
Kawasaki disease (KD) is an inflammatory disease in children associated with vasculitis affecting predominantly the coronary arteries and is now the most common cause of acquired heart disease in children in developed countries. The etiology of KD is unknown but epidemiological studies implicate an infectious agent or toxin, which causes disease in genetically predisposed individuals. The presence of immune complexes (ICs) in the serum of children with KD was established in numerous studies during the 1970s and 80s. More recent genetic studies have identified variation in Fcγ receptors and genes controlling immunoglobulin production associated with KD. In this review we link the genetic findings and IC studies and suggest a key role for their interaction in pathophysiology of the disease.
Collapse
Affiliation(s)
- Stephanie Menikou
- Section of Paediatrics, Division of Infectious Diseases, Department of Medicine, Imperial College London, London, United Kingdom
| | - Paul R Langford
- Section of Paediatrics, Division of Infectious Diseases, Department of Medicine, Imperial College London, London, United Kingdom
| | - Michael Levin
- Section of Paediatrics, Division of Infectious Diseases, Department of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
139
|
Abstract
Several pieces of evidence support the role of activated platelets in the development of the chronic inflammation-related diseases, such as atherothrombosis and cancer, mainly via the release of soluble factors and microparticles (MPs). Platelets and MPs contain a repertoire of proteins and genetic material (i.e., mRNAs and microRNAs) which may be influenced by the clinical condition of the individuals. In fact, platelets are capable of up-taking proteins and genetic material during their lifespan. Moreover, the content of platelet-derived MPs can be delivered to other cells, including stromal, immune, epithelial, and cancer cells, to change their phenotype and functions, thus contributing to cancer promotion and its metastasization. Platelets and MPs can play an indirect role in the metastatic process by helping malignant cells to escape from immunological surveillance. Furthermore, platelets and their derived MPs represent a potential source for blood biomarker development in oncology. This review provides an updated overview of the roles played by platelets and MPs in cancer and metastasis formation. The possible analysis of platelet and MP molecular signatures for the detection of cancer and monitoring of anticancer treatments is discussed. Finally, the potential use of MPs as vectors for drug delivery systems to cancer cells is put forward.
Collapse
|
140
|
Nurden AT. Acquired Glanzmann thrombasthenia: From antibodies to anti-platelet drugs. Blood Rev 2019; 36:10-22. [PMID: 31010659 DOI: 10.1016/j.blre.2019.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/15/2019] [Accepted: 03/19/2019] [Indexed: 02/06/2023]
Abstract
In contrast to the inherited platelet disorder given by mutations in the ITGA2B and ITGB3 genes, mucocutaneous bleeding from a spontaneous inhibition of normally expressed αIIbβ3 characterizes acquired Glanzmann thrombasthenia (GT). Classically, it is associated with autoantibodies or paraproteins that block platelet aggregation without causing a fall in platelet count. However, inhibitory antibodies to αIIbβ3 are widely associated with primary immune thrombocytopenia (ITP), occur in secondary ITP associated with leukemia and related disorders, solid cancers and myeloma, other autoimmune diseases, following organ transplantation while cytoplasmic dysregulation of αIIbβ3 function features in myeloproliferative and myelodysplastic syndromes. Antibodies to αIIbβ3 occur during viral and bacterial infections, while drug-dependent antibodies reacting with αIIbβ3 are a special case. Direct induction of acquired GT is a feature of therapies that block platelets in coronary artery disease. This review looks at these conditions, emphasizing molecular mechanisms, therapy, patient management and future directions for research.
Collapse
Affiliation(s)
- Alan T Nurden
- Institut de Rhythmologie et de Modélisation Cardiaque, Plateforme Technologique d'Innovation Biomédicale, Hôpital Xavier Arnozan, Pessac, France.
| |
Collapse
|
141
|
Becker Y, Loignon RC, Julien AS, Marcoux G, Allaeys I, Lévesque T, Rollet-Labelle E, Benk-Fortin H, Cloutier N, Melki I, Eder L, Wagner É, Pelletier M, Hajj HE, Tremblay MÈ, Belleannée C, Hébert MJ, Dieudé M, Rauch J, Fortin PR, Boilard E. Anti-mitochondrial autoantibodies in systemic lupus erythematosus and their association with disease manifestations. Sci Rep 2019; 9:4530. [PMID: 30872710 PMCID: PMC6418244 DOI: 10.1038/s41598-019-40900-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/21/2019] [Indexed: 12/13/2022] Open
Abstract
Mitochondria are organelles that govern energy supply and control cell death. Mitochondria also express bacterial features, such as the presence of inner membrane cardiolipin and a circular genome rich in hypomethylated CpG motifs. While mitochondrial extrusion by damaged organs or activated cells is thought to trigger innate immunity, it is unclear whether extracellular mitochondria also stimulate an adaptive immune response. We describe the development of novel assays to detect autoantibodies specific to two distinct components of the mitochondrion: the mitochondrial outer membrane and mitochondrial DNA. Antibodies to these two mitochondrial constituents were increased in both human and murine systemic lupus erythematosus (SLE), compared to controls, and were present at higher levels than in patients with antiphospholipid syndrome or primary biliary cirrhosis. In both bi- and multi-variate regression models, antibodies to mitochondrial DNA, but not whole mitochondria, were associated with increased anti-dsDNA antibodies and lupus nephritis. This study describes new and optimized methods for the assessment of anti-mitochondrial antibodies, and demonstrates their presence in both human and murine SLE. These findings suggest that different mitochondrial components are immunogenic in SLE, and support the concept that extracellular mitochondria may provide an important source of circulating autoantigens in SLE.
Collapse
Affiliation(s)
- Yann Becker
- Centre de Recherche du CHU de Québec - Université Laval, Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Québec, Qc, Canada
| | - Renée-Claude Loignon
- Division de Rhumatologie, Département de Médecine, CHU de Québec - Université Laval, Québec, Qc, Canada
| | - Anne-Sophie Julien
- Département de mathématiques et statistique, Université Laval, Québec, Qc, Canada
| | - Geneviève Marcoux
- Centre de Recherche du CHU de Québec - Université Laval, Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Québec, Qc, Canada
| | - Isabelle Allaeys
- Centre de Recherche du CHU de Québec - Université Laval, Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Québec, Qc, Canada
| | - Tania Lévesque
- Centre de Recherche du CHU de Québec - Université Laval, Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Québec, Qc, Canada
| | - Emmanuelle Rollet-Labelle
- Centre de Recherche du CHU de Québec - Université Laval, Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Québec, Qc, Canada
| | - Hadrien Benk-Fortin
- Centre de Recherche du CHU de Québec - Université Laval, Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Québec, Qc, Canada
| | - Nathalie Cloutier
- Centre de Recherche du CHU de Québec - Université Laval, Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Québec, Qc, Canada
| | - Imène Melki
- Centre de Recherche du CHU de Québec - Université Laval, Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Québec, Qc, Canada
| | - Lihi Eder
- Women's College Hospital and University of Toronto, Toronto, ON, Canada
| | - Éric Wagner
- Immunology and Histocompatibility Laboratory, Department of Medical Biology CHU de Québec - Université Laval; Department of Microbiology, Infectious Diseases and Immunology, Université Laval, Québec, Qc, Canada
| | - Martin Pelletier
- Centre de Recherche du CHU de Québec - Université Laval, Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Québec, Qc, Canada.,Axe maladies infectieuses et inflammatoires, Centre de recherche du CHU de Québec - Université Laval, Québec, Qc, Canada
| | - Hassan El Hajj
- Axe Neurosciences, Centre de Recherche du CHU de Québec - Université Laval, Québec, Qc, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec - Université Laval, Québec, Qc, Canada
| | - Clémence Belleannée
- Axe Reproduction, Santé de la mère et de l'enfant, Centre de recherche du CHU de Québec - Université Laval, Québec, Qc, Canada
| | - Marie-Josée Hébert
- Centre de recherche du CHU de Montréal, Montréal Québec, Québec, Qc, Canada
| | - Mélanie Dieudé
- Centre de recherche du CHU de Montréal, Montréal Québec, Québec, Qc, Canada
| | - Joyce Rauch
- Division of Rheumatology, Department of Medicine, Research Institute of the McGill University Health Centre, Montreal, Qc, H4A 3J1, Canada
| | - Paul R Fortin
- Division de Rhumatologie, Département de Médecine, CHU de Québec - Université Laval, Québec, Qc, Canada. .,Axe maladies infectieuses et inflammatoires, Centre de recherche du CHU de Québec - Université Laval, Québec, Qc, Canada.
| | - Eric Boilard
- Centre de Recherche du CHU de Québec - Université Laval, Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Québec, Qc, Canada. .,Axe maladies infectieuses et inflammatoires, Centre de recherche du CHU de Québec - Université Laval, Québec, Qc, Canada.
| |
Collapse
|
142
|
Abstract
Inflammatory arthritis encompasses a set of common diseases characterized by immune-mediated attack on joint tissues. Most but not all affected patients manifest circulating autoantibodies. Decades of study in human and animal arthritis have identified key roles for autoantibodies in immune complexes and through direct modulation of articular biology. However, joint inflammation can arise because of pathogenic T cells and other pathways that are antibody-independent. Here we review the evidence for these parallel tracks, in animal models and in humans, to explore the range of mechanisms engaged in the pathophysiology of arthritis and to highlight opportunities for targeted therapeutic intervention.
Collapse
Affiliation(s)
- Margaret H. Chang
- Department of Medicine, Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Peter A. Nigrovic
- Department of Medicine, Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
143
|
Liu Z, Zeng Z, Wu C, Liu H. Tropisetron inhibits sepsis by repressing hyper-inflammation and regulating the cardiac action potential in rat models. Biomed Pharmacother 2019; 110:380-388. [PMID: 30529771 DOI: 10.1016/j.biopha.2018.11.142] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/22/2018] [Accepted: 11/28/2018] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE The objective of the present investigation was to explore the possible effect of the 5-HT3 receptor antagonist tropisetron on the expression levels of the inflammatory factors interleukin 6 (IL-6), creatine kinase isoenzyme (CK-MB), soluble growth stimulating gene 2 protein (sST2) and immunoglobulin E (IgE), as well as the cardiac action potential in septic rats. METHODS The cecal ligation and perforation (CLP) method was utilized to construct abdominal infarction in rats. A total of 68 male adult Sprague Dawley rats were used, including 40 for assessing survival and 28 for detecting the expression levels of IL-6 and IgE, myocardial injury, cardiac dysfunction and the cardiac action potential. These 28 rats were divided into the sham (6 rats), sham + Tropisetron (6 rats), CLP (8 rats) and CLP + Tropisetron (8 rats) groups. Twenty-four hours after establishment of the sepsis rat model, immunohistochemistry was used to analyze 5-HT3 receptor protein expression, and enzyme-linked immunosorbent assay (ELISA) was employed to monitor the serum levels of IL-6, CKMB, sST2 and IgE. Furthermore, the structure of the myocardium in various groups was examined by H&E staining. RESULTS The levels of IL-6, CK-MB, sST2 and IgE in the sepsis group were significantly higher than those of the sham group (P < 0.01). Furthermore, the heart rate in the sepsis group was lower than that of the sham group (P < 0.01), and the time of atrial ventricular action potential in the sepsis group was longer than that of the sham group (P < 0.05). In addition, immunohistochemical analyses showed that the area, intensity and index of 5-HT3 receptor in the sepsis group were significantly lower than those of the sham group (P < 0.01). Importantly, the 5-HT3 receptor antagonist Tropisetron exhibited significant inhibitory effects IL-6, CK-MB, sST2 and IgE expression levels, and inductive effects on atrial ventricular action potential in the sepsis group. CONCLUSIONS Sepsis leads to systemic inflammatory reaction, resulting in myocardial injury, structural changes and immune imbalance. The inhibitory effect of tropisetron on inflammation, and the regulatory inflammatory disorder by the efferent vagus nerve may be one of the important mechanisms leading to cardiac electrophysiological changes in sepsis.
Collapse
Affiliation(s)
- Zhengjiang Liu
- Department of Cardiology, the Six Affiliated Hospital of Guangzhou Medical University/ Qingyuan People's Hospital, Qingyuan 511500, China.
| | - Zhiheng Zeng
- Department of Cardiology, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Changdong Wu
- NO.1 Deppartment of ICU, the People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830000, China
| | - Hua Liu
- Department of Cardiology, the Six Affiliated Hospital of Guangzhou Medical University/ Qingyuan People's Hospital, Qingyuan 511500, China
| |
Collapse
|
144
|
Liposomes with cyclic RGD peptide motif triggers acute immune response in mice. J Control Release 2019; 293:201-214. [DOI: 10.1016/j.jconrel.2018.12.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/18/2018] [Accepted: 12/03/2018] [Indexed: 12/25/2022]
|
145
|
|
146
|
The Role of Platelets in Antimicrobial Host Defense. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00029-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
|
147
|
Claushuis TAM, de Vos AF, Roelofs JJTH, de Boer OJ, van 't Veer C, van der Poll T. Platelet-Dense Granules Worsen Pre-Infection Thrombocytopenia during Gram-Negative Pneumonia-Derived Sepsis. J Innate Immun 2018; 11:168-180. [PMID: 30557883 DOI: 10.1159/000494147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 09/23/2018] [Indexed: 12/12/2022] Open
Abstract
Platelet-dense (δ) granules are important for platelet function. Platelets contribute to host defense and vascular integrity during pneumonia and sepsis, and δ granule products, including adenosine diphosphate (ADP), can influence inflammatory responses. We therefore aimed to study the role of platelet δ granules in the host response during sepsis. Hermansky-Pudlak syndrome (Hps)3coa mice (with reduced δ granule content), mice treated with the platelet ADP receptor inhibitor clopidogrel, and appropriate control mice were infected with the human sepsis pathogen Klebsiella pneumoniae via the airways to induce pneumonia and sepsis. In order to override potential redundancy in platelet functions, we also studied Hps3coa and control mice with moderate antibody-induced thrombocytopenia (10%) prior to infection. We found that sepsis-induced thrombocytopenia tended to be less severe in Hps3coa mice, and was significantly ameliorated in Hps3coa mice with low pre-infection platelet counts. Bacterial growth was similar in Hps3coa and control mice in the presence of normal platelet counts prior to infection, but lower in the lungs of Hps3coa mice with low pre-infection platelet counts. Hps3coa mice had unaltered lung pathology and distant organ injury during pneumosepsis, irrespective of pre-infection platelet counts; lung bleeding did not differ between Hps3coa and control mice. Clopidogrel did not influence any host response parameter. These data suggest that platelet δ granules can play a detrimental role in pneumosepsis by aggravating thrombocytopenia and impairing local antibacterial defense, but that these unfavorable effects only become apparent in the presence of low platelet counts.
Collapse
Affiliation(s)
- Theodora A M Claushuis
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands,
| | - Alex F de Vos
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Onno J de Boer
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Cornelis van 't Veer
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
148
|
Morrell CN, Pariser DN, Hilt ZT, Vega Ocasio D. The Platelet Napoleon Complex-Small Cells, but Big Immune Regulatory Functions. Annu Rev Immunol 2018; 37:125-144. [PMID: 30485751 DOI: 10.1146/annurev-immunol-042718-041607] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Platelets have dual physiologic roles as both cellular mediators of thrombosis and immune modulatory cells. Historically, the thrombotic function of platelets has received significant research and clinical attention, but emerging research indicates that the immune regulatory roles of platelets may be just as important. We now know that in addition to their role in the acute thrombotic event at the time of myocardial infarction, platelets initiate and accelerate inflammatory processes that are part of the pathogenesis of atherosclerosis and myocardial infarction expansion. Furthermore, it is increasingly apparent from recent studies that platelets impact the pathogenesis of many vascular inflammatory processes such as autoimmune diseases, sepsis, viral infections, and growth and metastasis of many types of tumors. Therefore, we must consider platelets as immune cells that affect all phases of immune responses.
Collapse
Affiliation(s)
- Craig N Morrell
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Rochester, New York 14642, USA;
| | - Daphne N Pariser
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Rochester, New York 14642, USA;
| | - Zachary T Hilt
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Rochester, New York 14642, USA;
| | - Denisse Vega Ocasio
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine, Rochester, New York 14642, USA;
| |
Collapse
|
149
|
Eisinger F, Patzelt J, Langer HF. The Platelet Response to Tissue Injury. Front Med (Lausanne) 2018; 5:317. [PMID: 30483508 PMCID: PMC6242949 DOI: 10.3389/fmed.2018.00317] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 10/23/2018] [Indexed: 12/16/2022] Open
Abstract
In recent years, various studies have increasingly explained platelet functions not only in their central role as a regulator in cellular hemostasis and coagulation. In fact, there is growing evidence that under specific conditions, platelets act as a mediator between the vascular system, hemostasis, and the immune system. Therefore, they are essential in many processes involved in tissue remodeling and tissue reorganization after injury or inflammatory responses. These processes include the promotion of inflammatory processes, the contribution to innate and adaptive immune responses during bacterial and viral infections, the modulation of angiogenesis, and the regulation of cell apoptosis in steady-state tissue homeostasis or after tissue breakdown. All in all platelets may contribute to the control of tissue homeostasis much more than generally assumed. This review summarizes the current knowledge of platelets as part of the tissue remodeling network and seeks to provide possible translational implications for clinical therapy.
Collapse
Affiliation(s)
- Felix Eisinger
- Section for Cardioimmunology, Department of Cardiovascular Medicine, University of Tuebingen, Tübingen, Germany
| | - Johannes Patzelt
- University Clinic for Cardiovascular Medicine, University of Tuebingen, Tübingen, Germany
| | - Harald F. Langer
- Section for Cardioimmunology, Department of Cardiovascular Medicine, University of Tuebingen, Tübingen, Germany
- University Clinic for Cardiovascular Medicine, University of Tuebingen, Tübingen, Germany
| |
Collapse
|
150
|
Platelet communication with the vascular wall: role of platelet-derived microparticles and non-coding RNAs. Clin Sci (Lond) 2018; 132:1875-1888. [PMID: 30185611 DOI: 10.1042/cs20180580] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 07/31/2018] [Accepted: 08/09/2018] [Indexed: 12/24/2022]
Abstract
Platelets play an important role in vascular homeostasis through their interaction with circulating blood cells as well as the vascular wall. Platelet-mediated communication with other cells can take the form of direct cell-cell interactions via membrane receptors or indirectly through the release of different soluble factors stored in their granules as well as through the release of microparticles. The latter carry different proteins and RNAs which are transferred to the target cells. The aim of this review is to discuss the role of platelet-derived factors, adhesion molecules as well as RNAs as mediators of the cross-talk between platelets and the vessel wall.
Collapse
|