1
|
Hawwari I, Rossnagel L, Rosero N, Maasewerd S, Vasconcelos MB, Jentzsch M, Demczuk A, Teichmann LL, Meffert L, Bertheloot D, Ribeiro LS, Kallabis S, Meissner F, Arditi M, Atici AE, Noval Rivas M, Franklin BS. Platelet transcription factors license the pro-inflammatory cytokine response of human monocytes. EMBO Mol Med 2024; 16:1901-1929. [PMID: 38977927 PMCID: PMC11319489 DOI: 10.1038/s44321-024-00093-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 07/10/2024] Open
Abstract
In humans, blood Classical CD14+ monocytes contribute to host defense by secreting large amounts of pro-inflammatory cytokines. Their aberrant activity causes hyper-inflammation and life-threatening cytokine storms, while dysfunctional monocytes are associated with 'immunoparalysis', a state of immune hypo responsiveness and reduced pro-inflammatory gene expression, predisposing individuals to opportunistic infections. Understanding how monocyte functions are regulated is critical to prevent these harmful outcomes. We reveal platelets' vital role in the pro-inflammatory cytokine responses of human monocytes. Naturally low platelet counts in patients with immune thrombocytopenia or removal of platelets from healthy monocytes result in monocyte immunoparalysis, marked by impaired cytokine response to immune challenge and weakened host defense transcriptional programs. Remarkably, supplementing monocytes with fresh platelets reverses these conditions. We discovered that platelets serve as reservoirs of key cytokine transcription regulators, such as NF-κB and MAPK p38, and pinpointed the enrichment of platelet NF-κB2 in human monocytes by proteomics. Platelets proportionally restore impaired cytokine production in human monocytes lacking MAPK p38α, NF-κB p65, and NF-κB2. We uncovered a vesicle-mediated platelet-monocyte-propagation of inflammatory transcription regulators, positioning platelets as central checkpoints in monocyte inflammation.
Collapse
Affiliation(s)
- Ibrahim Hawwari
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany.
| | - Lukas Rossnagel
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Nathalia Rosero
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Salie Maasewerd
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | | | - Marius Jentzsch
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Agnieszka Demczuk
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lino L Teichmann
- Department of Medicine III, University Hospital Bonn, Bonn, Germany
| | - Lisa Meffert
- Department of Medicine III, University Hospital Bonn, Bonn, Germany
| | - Damien Bertheloot
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lucas S Ribeiro
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Sebastian Kallabis
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Felix Meissner
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Moshe Arditi
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Asli E Atici
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Bernardo S Franklin
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany.
| |
Collapse
|
2
|
Noval Rivas M, Kocatürk B, Franklin BS, Arditi M. Platelets in Kawasaki disease: mediators of vascular inflammation. Nat Rev Rheumatol 2024; 20:459-472. [PMID: 38886559 DOI: 10.1038/s41584-024-01119-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 06/20/2024]
Abstract
Kawasaki disease, a systemic vasculitis that affects young children and can result in coronary artery aneurysms, is the leading cause of acquired heart disease among children. A hallmark of Kawasaki disease is increased blood platelet counts and platelet activation, which is associated with an increased risk of developing resistance to intravenous immunoglobulin and coronary artery aneurysms. Platelets and their releasate, including granules, microparticles, microRNAs and transcription factors, can influence innate immunity, enhance inflammation and contribute to vascular remodelling. Growing evidence indicates that platelets also interact with immune and non-immune cells to regulate inflammation. Platelets boost NLRP3 inflammasome activation and IL-1β production by human immune cells by releasing soluble mediators. Activated platelets form aggregates with leukocytes, such as monocytes and neutrophils, enhancing numerous functions of these cells and promoting thrombosis and inflammation. Leukocyte-platelet aggregates are increased in children with Kawasaki disease during the acute phase of the disease and can be used as biomarkers for disease severity. Here we review the role of platelets in Kawasaki disease and discuss progress in understanding the immune-effector role of platelets in amplifying inflammation related to Kawasaki disease vasculitis and therapeutic strategies targeting platelets or platelet-derived molecules.
Collapse
Affiliation(s)
- Magali Noval Rivas
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Begüm Kocatürk
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Bernardo S Franklin
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Moshe Arditi
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA.
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Gong Q, Fu M, Wang J, Zhao S, Wang H. Potential Immune-Inflammatory Proteome Biomarkers for Guiding the Treatment of Patients with Primary Acute Angle-Closure Glaucoma Caused by COVID-19. J Proteome Res 2024; 23:2587-2597. [PMID: 38836775 PMCID: PMC11232099 DOI: 10.1021/acs.jproteome.4c00325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/06/2024]
Abstract
Primary acute angle-closure glaucoma (PAACG) is a sight-threatening condition that can lead to blindness. With the increasing incidence of COVID-19, a multitude of people are experiencing acute vision loss and severe swelling of the eyes and head. These patients were then diagnosed with acute angle closure, with or without a history of PACG. However, the mechanism by which viral infection causes PACG has not been clarified. This is the first study to explore the specific inflammatory proteomic landscape in SARS-CoV-2-induced PAACG. The expression of 92 inflammation-related proteins in 19 aqueous humor samples from PAACGs or cataract patients was detected using the Olink Target 96 Inflammation Panel based on a highly sensitive and specific proximity extension assay technology. The results showed that 76 proteins were significantly more abundant in the PAACG group than in the cataract group. Notably, the top eight differentially expressed proteins were IL-8, MCP-1, TNFRSF9, DNER, CCL4, Flt3L, CXCL10, and CD40. Generally, immune markers are related to inflammation, macrophage activation, and viral infection, revealing the crucial role of macrophages in the occurrence of PAACGs caused by SARS-CoV-2.
Collapse
Affiliation(s)
- Qiaoyun Gong
- Department
of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
- National
Clinical Research Center for Eye Diseases, Shanghai 200080, China
- Shanghai
Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
- Shanghai
Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
- Shanghai
Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai 200080, China
| | - Mingshui Fu
- Department
of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
- National
Clinical Research Center for Eye Diseases, Shanghai 200080, China
- Shanghai
Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
- Shanghai
Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
- Shanghai
Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai 200080, China
| | - Jingyi Wang
- Department
of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
- National
Clinical Research Center for Eye Diseases, Shanghai 200080, China
- Shanghai
Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
- Shanghai
Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
- Shanghai
Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai 200080, China
| | - Shuzhi Zhao
- Department
of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
- National
Clinical Research Center for Eye Diseases, Shanghai 200080, China
- Shanghai
Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
- Shanghai
Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
- Shanghai
Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai 200080, China
| | - Haiyan Wang
- Department
of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China
- National
Clinical Research Center for Eye Diseases, Shanghai 200080, China
- Shanghai
Key Laboratory of Ocular Fundus Diseases, Shanghai 200080, China
- Shanghai
Engineering Center for Visual Science and Photomedicine, Shanghai 200080, China
- Shanghai
Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai 200080, China
| |
Collapse
|
4
|
Mack A, Vanden Hoek T, Du X. Thromboinflammation and the Role of Platelets. Arterioscler Thromb Vasc Biol 2024; 44:1175-1180. [PMID: 38776384 DOI: 10.1161/atvbaha.124.320149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Affiliation(s)
- Andrew Mack
- Department of Pharmacology and Regenerative Medicine (A.M., X.D.), University of Illinois, Chicago
| | - Terry Vanden Hoek
- Department of Emergency Medicine (T.V.H.), University of Illinois, Chicago
| | - Xiaoping Du
- Department of Pharmacology and Regenerative Medicine (A.M., X.D.), University of Illinois, Chicago
| |
Collapse
|
5
|
Qiu X, Nair MG, Jaroszewski L, Godzik A. Deciphering Abnormal Platelet Subpopulations in COVID-19, Sepsis and Systemic Lupus Erythematosus through Machine Learning and Single-Cell Transcriptomics. Int J Mol Sci 2024; 25:5941. [PMID: 38892129 PMCID: PMC11173046 DOI: 10.3390/ijms25115941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
This study focuses on understanding the transcriptional heterogeneity of activated platelets and its impact on diseases such as sepsis, COVID-19, and systemic lupus erythematosus (SLE). Recognizing the limited knowledge in this area, our research aims to dissect the complex transcriptional profiles of activated platelets to aid in developing targeted therapies for abnormal and pathogenic platelet subtypes. We analyzed single-cell transcriptional profiles from 47,977 platelets derived from 413 samples of patients with these diseases, utilizing Deep Neural Network (DNN) and eXtreme Gradient Boosting (XGB) to distinguish transcriptomic signatures predictive of fatal or survival outcomes. Our approach included source data annotations and platelet markers, along with SingleR and Seurat for comprehensive profiling. Additionally, we employed Uniform Manifold Approximation and Projection (UMAP) for effective dimensionality reduction and visualization, aiding in the identification of various platelet subtypes and their relation to disease severity and patient outcomes. Our results highlighted distinct platelet subpopulations that correlate with disease severity, revealing that changes in platelet transcription patterns can intensify endotheliopathy, increasing the risk of coagulation in fatal cases. Moreover, these changes may impact lymphocyte function, indicating a more extensive role for platelets in inflammatory and immune responses. This study identifies crucial biomarkers of platelet heterogeneity in serious health conditions, paving the way for innovative therapeutic approaches targeting platelet activation, which could improve patient outcomes in diseases characterized by altered platelet function.
Collapse
Affiliation(s)
| | | | | | - Adam Godzik
- Division of Biomedical Sciences, University of California Riverside School of Medicine, Riverside, CA 92521, USA; (X.Q.); (M.G.N.); (L.J.)
| |
Collapse
|
6
|
Nicolai L, Pekayvaz K, Massberg S. Platelets: Orchestrators of immunity in host defense and beyond. Immunity 2024; 57:957-972. [PMID: 38749398 DOI: 10.1016/j.immuni.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 04/06/2024] [Accepted: 04/12/2024] [Indexed: 06/05/2024]
Abstract
Platelets prevent blood loss during vascular injury and contribute to thrombus formation in cardiovascular disease. Beyond these classical roles, platelets are critical for the host immune response. They guard the vasculature against pathogens via specialized receptors, intracellular signaling cascades, and effector functions. Platelets also skew inflammatory responses by instructing innate immune cells, support adaptive immunosurveillance, and influence antibody production and T cell polarization. Concomitantly, platelets contribute to tissue reconstitution and maintain vascular function after inflammatory challenges. However, dysregulated activation of these multitalented cells exacerbates immunopathology with ensuing microvascular clotting, excessive inflammation, and elevated risk of macrovascular thrombosis. This dichotomy underscores the critical importance of precisely defining and potentially modulating platelet function in immunity.
Collapse
Affiliation(s)
- Leo Nicolai
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Kami Pekayvaz
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, University Hospital Ludwig-Maximilian University, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
7
|
Luo S, Xu R, Xie P, Liu X, Ling C, Liu Y, Zhang X, Xia Z, Chen Z, Tang J. EGFR of platelet regulates macrophage activation and bacterial phagocytosis function. J Inflamm (Lond) 2024; 21:10. [PMID: 38632608 PMCID: PMC11022435 DOI: 10.1186/s12950-024-00382-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/03/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Beyond their crucial role in hemostasis, platelets possess the ability to regulate inflammation and combat infections through various mechanisms. Stringent control of macrophage activation is essential during innate immune responses in sepsis. Macrophages are considered crucial phagocytic cells that aid in the elimination of pathogens. Platelet interactions with monocytes-macrophages are known to be significant in the response against bacterial infections, but the primary mediator driving these interactions remains unclear. EGFR plays critical role in the regulation of inflammation and infection through various mechanisms. RESULTS The overexpression of platelets by thrombopoietin (TPO) leads to the sequestration of both pro-inflammatory (IL-6/IL-1) and anti-inflammatory (IL-10) cytokines in the organ tissue of septic mice. Epidermal growth factor receptor (EGFR) is critical for platelet activation in sepsis. EGFR-licensed platelets enhance macrophage immune function, including the production of reactive oxygen species (ROS) and the clearance of bacteria. Platelet EGFR also induces M1 macrophage polarization by increasing the expression of inducible nitric oxide synthase (iNOS) and CD64. CONCLUSION EGFR can activate platelet immune function. Moreover, activated platelets efficiently regulate bacterial phagocytosis and pro-inflammatory function of macrophages through an EGFR-dependent pathway.
Collapse
Affiliation(s)
- Shuhua Luo
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524000, Zhanjiang, Guangdong, China
- Guang Dong Medical University, 524000, Zhanjiang, Guangdong, China
| | - Riping Xu
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524000, Zhanjiang, Guangdong, China
| | - Pengyun Xie
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524000, Zhanjiang, Guangdong, China
| | - Xiaolei Liu
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524000, Zhanjiang, Guangdong, China
- Guang Dong Medical University, 524000, Zhanjiang, Guangdong, China
| | - Chunxiu Ling
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524000, Zhanjiang, Guangdong, China
- Guang Dong Medical University, 524000, Zhanjiang, Guangdong, China
| | - Yusha Liu
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524000, Zhanjiang, Guangdong, China
- Guang Dong Medical University, 524000, Zhanjiang, Guangdong, China
| | - Xuedi Zhang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524000, Zhanjiang, Guangdong, China
- Guang Dong Medical University, 524000, Zhanjiang, Guangdong, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524000, Zhanjiang, Guangdong, China
| | - Zhanghui Chen
- Zhanjiang Institute of Clinical Medicine, Zhanjiang Central Hospital, Guangdong Medical University, 524000, Zhanjiang, Guangdong, China.
| | - Jing Tang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, 524000, Zhanjiang, Guangdong, China.
| |
Collapse
|
8
|
Li C, Ture SK, Nieves-Lopez B, Blick-Nitko SK, Maurya P, Livada AC, Stahl TJ, Kim M, Pietropaoli AP, Morrell CN. Thrombocytopenia Independently Leads to Changes in Monocyte Immune Function. Circ Res 2024; 134:970-986. [PMID: 38456277 PMCID: PMC11069346 DOI: 10.1161/circresaha.123.323662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND While platelets have well-studied hemostatic functions, platelets are immune cells that circulate at the interface between the vascular wall and white blood cells. The physiological implications of these constant transient interactions are poorly understood. Activated platelets induce and amplify immune responses, but platelets may also maintain immune homeostasis in healthy conditions, including maintaining vascular integrity and T helper cell differentiation, meaning that platelets are central to both immune responses and immune quiescence. Clinical data have shown an association between low platelet counts (thrombocytopenia) and immune dysfunction in patients with sepsis and extracorporeal membrane oxygenation, further implicating platelets as more holistic immune regulators, but studies of platelet immune functions in nondisease contexts have had limited study. METHODS We used in vivo models of thrombocytopenia and in vitro models of platelet and monocyte interactions, as well as RNA-seq and ATAC-seq (assay for transposase-accessible chromatin with sequencing), to mechanistically determine how resting platelet and monocyte interactions immune program monocytes. RESULTS Circulating platelets and monocytes interact in a CD47-dependent manner to regulate monocyte metabolism, histone methylation, and gene expression. Resting platelet-monocyte interactions limit TLR (toll-like receptor) signaling responses in healthy conditions in an innate immune training-like manner. In both human patients with sepsis and mouse sepsis models, thrombocytopenia exacerbated monocyte immune dysfunction, including increased cytokine production. CONCLUSIONS Thrombocytopenia immune programs monocytes in a manner that may lead to immune dysfunction in the context of sepsis. This is the first demonstration that sterile, endogenous cell interactions between resting platelets and monocytes regulate monocyte metabolism and pathogen responses, demonstrating platelets to be immune rheostats in both health and disease.
Collapse
Affiliation(s)
- Chen Li
- Aab Cardiovascular Research Institute (C.L., S.K.T., B.N.-L., S.K.B.-N., P.M., A.C.L., C.N.M.), University of Rochester School of Medicine and Dentistry, NY
| | - Sara K Ture
- Aab Cardiovascular Research Institute (C.L., S.K.T., B.N.-L., S.K.B.-N., P.M., A.C.L., C.N.M.), University of Rochester School of Medicine and Dentistry, NY
| | - Benjamin Nieves-Lopez
- Aab Cardiovascular Research Institute (C.L., S.K.T., B.N.-L., S.K.B.-N., P.M., A.C.L., C.N.M.), University of Rochester School of Medicine and Dentistry, NY
- University of Puerto Rico, Medical Sciences Campus, San Juan (B.N.-L.)
| | - Sara K Blick-Nitko
- Aab Cardiovascular Research Institute (C.L., S.K.T., B.N.-L., S.K.B.-N., P.M., A.C.L., C.N.M.), University of Rochester School of Medicine and Dentistry, NY
| | - Preeti Maurya
- Aab Cardiovascular Research Institute (C.L., S.K.T., B.N.-L., S.K.B.-N., P.M., A.C.L., C.N.M.), University of Rochester School of Medicine and Dentistry, NY
| | - Alison C Livada
- Aab Cardiovascular Research Institute (C.L., S.K.T., B.N.-L., S.K.B.-N., P.M., A.C.L., C.N.M.), University of Rochester School of Medicine and Dentistry, NY
| | - Tyler J Stahl
- Genomics Research Center (T.J.S.), University of Rochester School of Medicine and Dentistry, NY
| | - Minsoo Kim
- Department of Microbiology and Immunology (M.K., C.N.M.), University of Rochester School of Medicine and Dentistry, NY
| | - Anthony P Pietropaoli
- Department of Medicine (A.P.P., C.N.M.), University of Rochester School of Medicine and Dentistry, NY
| | - Craig N Morrell
- Aab Cardiovascular Research Institute (C.L., S.K.T., B.N.-L., S.K.B.-N., P.M., A.C.L., C.N.M.), University of Rochester School of Medicine and Dentistry, NY
- Department of Microbiology and Immunology (M.K., C.N.M.), University of Rochester School of Medicine and Dentistry, NY
- Department of Medicine (A.P.P., C.N.M.), University of Rochester School of Medicine and Dentistry, NY
- Department of Pathology and Laboratory Medicine (C.N.M.), University of Rochester School of Medicine and Dentistry, NY
| |
Collapse
|
9
|
Vnučák M, Graňák K, Beliančinová M, Kleinová P, Blichová T, Doboš V, Dedinská I. Effect of Different Induction Immunosuppression on the Incidence of Infectious Complications after Kidney Transplantation-Single Center Study. J Clin Med 2024; 13:2162. [PMID: 38673435 PMCID: PMC11050246 DOI: 10.3390/jcm13082162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/29/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
Background/Objectives: Potent immunosuppression lowers the incidence of acute graft rejection but increases the risk of infections. In order to decrease either infectious complications or acute rejection, it is necessary to identify risk groups of patients profiting from personalized induction immunosuppressive treatment. The aim of our analysis was to find whether there were higher incidences of infectious complications after kidney transplantation (KT) in groups with different induction immunosuppressive treatment and also to find independent risk factors for recurrent infections. Materials: We retrospectively evaluated all patients with induction treatment with basiliximab after kidney transplantation from 2014 to 2019 at our center relative to age- and sex-matched controls of patients with thymoglobulin induction immunosuppression. Results: Our study consisted of two groups: basiliximab (39) and thymoglobulin (39). In the thymoglobulin group we observed an increased incidence of recurrent infection in every observed interval; however, acute rejection was seen more often in the basiliximab group. A history of respiratory diseases and thrombocytopenia were identified as independent risk factors for recurrent bacterial infections from the first to sixth month after KT. Decreased eGFR from the first month, infections caused by multi-drug-resistant bacteria, and severe infections (reflected by the need for hospitalization) were identified as independent risk factors for recurrent bacterial infections from the first to the twelfth month after KT. Conclusions: We found that in the group of patients with thymoglobulin induction immunosuppressive treatment, infectious complications occurred significantly more often during the entire monitored period with decreased incidence of acute humoral and cellular rejection occurred more often.
Collapse
Affiliation(s)
- Matej Vnučák
- Transplant-Nephrology Centre, University Hospital Martin, Kollárova 2, 03601 Martin, Slovakia; (M.V.); (M.B.); (P.K.); (T.B.); (I.D.)
- 1st Department of Internal Diseases, University Hospital Martin and Jessenius Medical Faculty of Comenius University, Kollárova 2, 03601 Martin, Slovakia;
| | - Karol Graňák
- Transplant-Nephrology Centre, University Hospital Martin, Kollárova 2, 03601 Martin, Slovakia; (M.V.); (M.B.); (P.K.); (T.B.); (I.D.)
- 1st Department of Internal Diseases, University Hospital Martin and Jessenius Medical Faculty of Comenius University, Kollárova 2, 03601 Martin, Slovakia;
| | - Monika Beliančinová
- Transplant-Nephrology Centre, University Hospital Martin, Kollárova 2, 03601 Martin, Slovakia; (M.V.); (M.B.); (P.K.); (T.B.); (I.D.)
- 1st Department of Internal Diseases, University Hospital Martin and Jessenius Medical Faculty of Comenius University, Kollárova 2, 03601 Martin, Slovakia;
| | - Patrícia Kleinová
- Transplant-Nephrology Centre, University Hospital Martin, Kollárova 2, 03601 Martin, Slovakia; (M.V.); (M.B.); (P.K.); (T.B.); (I.D.)
- 1st Department of Internal Diseases, University Hospital Martin and Jessenius Medical Faculty of Comenius University, Kollárova 2, 03601 Martin, Slovakia;
| | - Tímea Blichová
- Transplant-Nephrology Centre, University Hospital Martin, Kollárova 2, 03601 Martin, Slovakia; (M.V.); (M.B.); (P.K.); (T.B.); (I.D.)
- 1st Department of Internal Diseases, University Hospital Martin and Jessenius Medical Faculty of Comenius University, Kollárova 2, 03601 Martin, Slovakia;
| | - Vladimír Doboš
- 1st Department of Internal Diseases, University Hospital Martin and Jessenius Medical Faculty of Comenius University, Kollárova 2, 03601 Martin, Slovakia;
| | - Ivana Dedinská
- Transplant-Nephrology Centre, University Hospital Martin, Kollárova 2, 03601 Martin, Slovakia; (M.V.); (M.B.); (P.K.); (T.B.); (I.D.)
- 1st Department of Internal Diseases, University Hospital Martin and Jessenius Medical Faculty of Comenius University, Kollárova 2, 03601 Martin, Slovakia;
| |
Collapse
|
10
|
Claeys W, Verhaege D, Van Imschoot G, Van Wonterghem E, Van Acker L, Amelinck L, De Ponti FF, Scott C, Geerts A, Van Steenkiste C, Van Hoecke L, Vandenbroucke RE. Limitations of PLX3397 as a microglial investigational tool: peripheral and off-target effects dictate the response to inflammation. Front Immunol 2023; 14:1283711. [PMID: 38077359 PMCID: PMC10703484 DOI: 10.3389/fimmu.2023.1283711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 10/23/2023] [Indexed: 12/18/2023] Open
Abstract
Microglia, the resident macrophages of the central nervous system (CNS), play a critical role in CNS homeostasis and neuroinflammation. Pexidartinib (PLX3397), a colony-stimulating factor 1 (CSF1) receptor inhibitor, is widely used to deplete microglia, offering flexible options for both long-term depletion and highly versatile depletion-repopulation cycles. However, the potential impact of PLX3397 on peripheral (immune) cells remains controversial. Until now, the microglia-specificity of this type of compounds has not been thoroughly evaluated, particularly in the context of peripherally derived neuroinflammation. Our study addresses this gap by examining the effects of PLX3397 on immune cells in the brain, liver, circulation and bone marrow, both in homeostasis and systemic inflammation models. Intriguingly, we demonstrate that PLX3397 treatment not only influences the levels of tissue-resident macrophages, but also affects circulating and bone marrow immune cells beyond the mononuclear phagocyte system (MPS). These alterations in peripheral immune cells disrupt the response to systemic inflammation, consequently impacting the phenotype irrespective of microglial depletion. Furthermore, we observed that a lower dose of PLX3397, which does not deplete microglia, demonstrates similar (non-)MPS effects, both in the periphery and the brain, but fails to fully replicate the peripheral alterations seen in the higher doses, questioning lower doses as a 'peripheral control' strategy. Overall, our data highlight the need for caution when interpreting studies employing this compound, as it may not be suitable for specific investigation of microglial function in the presence of systemic inflammation.
Collapse
Affiliation(s)
- Wouter Claeys
- Department of Internal Medicine and Paediatrics, Hepatology Research Unit, Ghent University, Ghent, Belgium
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Daan Verhaege
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Griet Van Imschoot
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Elien Van Wonterghem
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Lore Van Acker
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Laura Amelinck
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Federico F. De Ponti
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB–UGent Center for Inflammation Research, Ghent, Belgium
| | - Charlotte Scott
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB–UGent Center for Inflammation Research, Ghent, Belgium
| | - Anja Geerts
- Department of Internal Medicine and Paediatrics, Hepatology Research Unit, Ghent University, Ghent, Belgium
- Liver Research Center Ghent, Ghent University, Ghent University Hospital, Ghent, Belgium
- Department of Gastroenterology and Hepatology, Ghent University Hospital, Ghent, Belgium
| | - Christophe Van Steenkiste
- Antwerp University, Department of Gastroenterology and Hepatology, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, Maria Middelares Hospital, Ghent, Belgium
| | - Lien Van Hoecke
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Roosmarijn E. Vandenbroucke
- Barriers in Inflammation, VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
11
|
Jiang J, Li W, Zhou L, Liu D, Wang Y, An J, Qiao S, Xie Z. Platelet ITGA2B inhibits caspase-8 and Rip3/Mlkl-dependent platelet death though PTPN6 during sepsis. iScience 2023; 26:107414. [PMID: 37554440 PMCID: PMC10404729 DOI: 10.1016/j.isci.2023.107414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/04/2023] [Accepted: 07/14/2023] [Indexed: 08/10/2023] Open
Abstract
Platelets play an important role in the pathogenesis of sepsis and platelet transfusion is a therapeutic option for sepsis patients, although the exact mechanisms have not been elucidated so far. ITGA2B encodes the αIIb protein in platelets, and its upregulation in sepsis is associated with increased mortality rate. Here, we generated a Itga2b (Q887X) knockin mouse, which significantly reduced ITGA2B expression of platelet and megakaryocyte. The decrease of ITGA2B level aggravated the death of septic mice. We analyzed the transcriptomic profiles of the platelets using RNA sequencing. Our findings suggest that ITGA2B upregulates PTPN6 in megakaryocytes via the transcription factors Nfkb1 and Rel. Furthermore, PTPN6 inhibits platelet apoptosis and necroptosis during sepsis by targeting the Ripk1/Ripk3/Mlkl and caspase-8 pathways. This prevents Kupffer cells from rapidly clearing activated platelets, and eventually maintains vascular integrity during sepsis. Our findings indicate a new function of ITGA2B in the regulation of platelet death during sepsis.
Collapse
Affiliation(s)
- Jiang Jiang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Li
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| | - Lu Zhou
- Hematology Department, Affiliated Hospital of Nantong University, Nantong, China
| | - Dengping Liu
- Suzhou Center for Disease Control and Prevention, Suzhou, China
| | - Yuanyuan Wang
- Department of Intensive Care Unit, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| | - Jianzhong An
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| | - Shigang Qiao
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
- Faculty of Anesthesiology, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| | - Zhanli Xie
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| |
Collapse
|
12
|
Yu S, Zhang Q, Su L, He J, Shi W, Yan H, Mao H, Sun Y, Cheng D, Wang X, Zhang Y, Fang L. Dabie bandavirus infection induces macrophagic pyroptosis and this process is attenuated by platelets. PLoS Negl Trop Dis 2023; 17:e0011488. [PMID: 37486928 PMCID: PMC10399884 DOI: 10.1371/journal.pntd.0011488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 06/29/2023] [Indexed: 07/26/2023] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is an emerging tick-borne infection with a high mortality rate in humans, which is caused by Dabie bandavirus (DBV), formerly known as SFTS virus. Clinical manifestations of SFTS are characterized by high fever, thrombocytopenia, leukopenia, hemorrhage, gastrointestinal symptoms, myalgia and local lymph node enlargement with up to 30% case fatality rates in human. Macrophage depletion in secondary lymphoid organs have important roles in the pathogenic process of fatal SFTS, but its exact cell death mechanism remains largely unknown. Here, we showed for the first time that DBV infection induced macrophagic pyroptosis, as evidenced by swollen cells, pore-forming structures, accumulation of gasdermin D N-terminal (GSDMD-NT) as well as the release of lactate dehydrogenase (LDH) and IL-1β in human macrophages. In addition to the upregulation of pyronecrosis genes, the expressions of pyroptosis-related proteins (GSDMD, caspase-1 and IL-1β) were also elevated. To be noted, platelets were found to play a protective role in DBV-derived pyroptosis. Transcriptome analysis and in vitro studies demonstrated that platelets significantly reduced the gene expressions and protein production of pro-pyroptotic markers and inflammatory cytokines in macrophages, whereas platelets conferred a propagation advantage for DBV. Collectively, this study demonstrates a novel mechanism by which DBV invasion triggers pyroptosis as a host defense to remove replication niches in human macrophages and platelets provide an additional layer to reduce cellular death. These findings may have important implications to the pathogenesis of lethal DBV, and provide new ideas for developing novel therapeutics to combat its infection.
Collapse
Affiliation(s)
- Sicong Yu
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
- The First People's Hospital of Xiaoshan District, Hangzhou, China
| | - Qinyi Zhang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Lingxuan Su
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Ji He
- Blood Center of Zhejiang Province, Hangzhou, China
| | - Wen Shi
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Hao Yan
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Haiyan Mao
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Yi Sun
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Dongqing Cheng
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuan Wang
- Shaoxing Shangyu District Center for Disease Control and Prevention, Shaoxing, China
| | - Yanjun Zhang
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Lei Fang
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| |
Collapse
|
13
|
Cai L, Rodgers E, Schoenmann N, Raju RP. Advances in Rodent Experimental Models of Sepsis. Int J Mol Sci 2023; 24:9578. [PMID: 37298529 PMCID: PMC10253762 DOI: 10.3390/ijms24119578] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/09/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
In the development of therapeutic strategies for human diseases, preclinical experimental models have a key role. However, the preclinical immunomodulatory therapies developed using rodent sepsis were not successful in human clinical trials. Sepsis is characterized by a dysregulated inflammation and redox imbalance triggered by infection. Human sepsis is simulated in experimental models using methods that trigger inflammation or infection in the host animals, most often mice or rats. It remains unknown whether the characteristics of the host species, the methods used to induce sepsis, or the molecular processes focused upon need to be revisited in the development of treatment methods that will succeed in human clinical trials. Our goal in this review is to provide a survey of existing experimental models of sepsis, including the use of humanized mice and dirty mice, and to show how these models reflect the clinical course of sepsis. We will discuss the strengths and limitations of these models and present recent advances in this subject area. We maintain that rodent models continue to have an irreplaceable role in studies toward discovering treatment methods for human sepsis.
Collapse
Affiliation(s)
- Lun Cai
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Elizabeth Rodgers
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Nick Schoenmann
- Department of Emergency Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Raghavan Pillai Raju
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
14
|
Jeong SH, Park JY, Ryu YB, Kim WS, Lee IC, Kim JH, Kim D, Ha JH, Lee BW, Nam J, Cho KO, Kwon HJ. Myristica fragrans Extract Inhibits Platelet Desialylation and Activation to Ameliorate Sepsis-Associated Thrombocytopenia in a Murine CLP-Induced Sepsis Model. Int J Mol Sci 2023; 24:ijms24108863. [PMID: 37240208 DOI: 10.3390/ijms24108863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Sepsis, characterized by an uncontrolled host inflammatory response to infections, remains a leading cause of death in critically ill patients worldwide. Sepsis-associated thrombocytopenia (SAT), a common disease in patients with sepsis, is an indicator of disease severity. Therefore, alleviating SAT is an important aspect of sepsis treatment; however, platelet transfusion is the only available treatment strategy for SAT. The pathogenesis of SAT involves increased platelet desialylation and activation. In this study, we investigated the effects of Myristica fragrans ethanol extract (MF) on sepsis and SAT. Desialylation and activation of platelets treated with sialidase and adenosine diphosphate (platelet agonist) were assessed using flow cytometry. The extract inhibited platelet desialylation and activation via inhibiting bacterial sialidase activity in washed platelets. Moreover, MF improved survival and reduced organ damage and inflammation in a mouse model of cecal ligation and puncture (CLP)-induced sepsis. It also prevented platelet desialylation and activation via inhibiting circulating sialidase activity, while maintaining platelet count. Inhibition of platelet desialylation reduces hepatic Ashwell-Morell receptor-mediated platelet clearance, thereby reducing hepatic JAK2/STAT3 phosphorylation and thrombopoietin mRNA expression. This study lays a foundation for the development of plant-derived therapeutics for sepsis and SAT and provides insights into sialidase-inhibition-based sepsis treatment strategies.
Collapse
Affiliation(s)
- Seong-Hun Jeong
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Ji-Young Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology, Jeongeup 53212, Republic of Korea
| | - Young Bae Ryu
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Woo Sik Kim
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - In-Chul Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology, Jeongeup 53212, Republic of Korea
| | - Ju-Hong Kim
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Dohoon Kim
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Ji-Hye Ha
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Ba-Wool Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Jiyoung Nam
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Kyoung-Oh Cho
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hyung-Jun Kwon
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
- Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology, Jeongeup 53212, Republic of Korea
| |
Collapse
|
15
|
Rolling CC, Barrett TJ, Berger JS. Platelet-monocyte aggregates: molecular mediators of thromboinflammation. Front Cardiovasc Med 2023; 10:960398. [PMID: 37255704 PMCID: PMC10225702 DOI: 10.3389/fcvm.2023.960398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 04/24/2023] [Indexed: 06/01/2023] Open
Abstract
Platelets, key facilitators of primary hemostasis and thrombosis, have emerged as crucial cellular mediators of innate immunity and inflammation. Exemplified by their ability to alter the phenotype and function of monocytes, activated platelets bind to circulating monocytes to form monocyte-platelet aggregates (MPA). The platelet-monocyte axis has emerged as a key mechanism connecting thrombosis and inflammation. MPA are elevated across the spectrum of inflammatory and autoimmune disorders, including cardiovascular disease, systemic lupus erythematosus (SLE), and COVID-19, and are positively associated with disease severity. These clinical disorders are all characterized by an increased risk of thromboembolic complications. Intriguingly, monocytes in contact with platelets become proinflammatory and procoagulant, highlighting that this interaction is a central element of thromboinflammation.
Collapse
Affiliation(s)
- Christina C. Rolling
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
- Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tessa J. Barrett
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Jeffrey S. Berger
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
16
|
Li C, Ture SK, Nieves-Lopez B, Blick-Nitko SK, Maurya P, Livada AC, Stahl TJ, Kim M, Pietropaoli AP, Morrell CN. Thrombocytopenia Independently Leads to Monocyte Immune Dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.10.540214. [PMID: 37214993 PMCID: PMC10197656 DOI: 10.1101/2023.05.10.540214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
In addition to their well-studied hemostatic functions, platelets are immune cells. Platelets circulate at the interface between the vascular wall and leukocytes, and transient platelet-leukocyte complexes are found in both healthy and disease states, positioning platelets to provide physiologic cues of vascular health and injury. Roles for activated platelets in inducing and amplifying immune responses have received an increasing amount of research attention, but our past studies also showed that normal platelet counts are needed in healthy conditions to maintain immune homeostasis. We have now found that thrombocytopenia (a low platelet count) leads to monocyte dysfunction, independent of the cause of thrombocytopenia, in a manner that is dependent on direct platelet-monocyte CD47 interactions that regulate monocyte immunometabolism and gene expression. Compared to monocytes from mice with normal platelet counts, monocytes from thrombocytopenic mice had increased toll-like receptor (TLR) responses, including increased IL-6 production. Furthermore, ex vivo co-incubation of resting platelets with platelet naïve bone marrow monocytes, induced monocyte metabolic programming and durable changes in TLR agonist responses. Assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-Seq) on monocytes from thrombocytopenic mice showed persistently open chromatin at LPS response genes and resting platelet interactions with monocytes induced histone methylation in a CD47 dependent manner. Using mouse models of thrombocytopenia and sepsis, normal platelet numbers were needed to limit monocyte immune dysregulation and IL6 expression in monocytes from human patients with sepsis also inversely correlated with patient platelet counts. Our studies demonstrate that in healthy conditions, resting platelets maintain monocyte immune tolerance by regulating monocyte immunometabolic processes that lead to epigenetic changes in TLR-related genes. This is also the first demonstration of sterile cell interactions that regulate of innate immune-metabolism and monocyte pathogen responses.
Collapse
|
17
|
Sharma S, Tyagi T, Antoniak S. Platelet in thrombo-inflammation: Unraveling new therapeutic targets. Front Immunol 2022; 13:1039843. [PMID: 36451834 PMCID: PMC9702553 DOI: 10.3389/fimmu.2022.1039843] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
In the broad range of human diseases, thrombo-inflammation appears as a clinical manifestation. Clinically, it is well characterized in context of superficial thrombophlebitis that is recognized as thrombosis and inflammation of superficial veins. However, it is more hazardous when developed in the microvasculature of injured/inflamed/infected tissues and organs. Several diseases like sepsis and ischemia-reperfusion can cause formation of microvascular thrombosis subsequently leading to thrombo-inflammation. Thrombo-inflammation can also occur in cases of antiphospholipid syndrome, preeclampsia, sickle cell disease, bacterial and viral infection. One of the major contributors to thrombo-inflammation is the loss of normal anti-thrombotic and anti-inflammatory potential of the endothelial cells of vasculature. This manifest itself in the form of dysregulation of the coagulation pathway and complement system, pathologic platelet activation, and increased recruitment of leukocyte within the microvasculature. The role of platelets in hemostasis and formation of thrombi under pathologic and non-pathologic conditions is well established. Platelets are anucleate cells known for their essential role in primary hemostasis and the coagulation pathway. In recent years, studies provide strong evidence for the critical involvement of platelets in inflammatory processes like acute ischemic stroke, and viral infections like Coronavirus disease 2019 (COVID-19). This has encouraged the researchers to investigate the contribution of platelets in the pathology of various thrombo-inflammatory diseases. The inhibition of platelet surface receptors or their intracellular signaling which mediate initial platelet activation and adhesion might prove to be suitable targets in thrombo-inflammatory disorders. Thus, the present review summarizes the concept and mechanism of platelet signaling and briefly discuss their role in sterile and non-sterile thrombo-inflammation, with the emphasis on role of platelets in COVID-19 induced thrombo-inflammation. The aim of this review is to summarize the recent developments in deciphering the role of the platelets in thrombo-inflammation and discuss their potential as pharmaceutical targets.
Collapse
Affiliation(s)
- Swati Sharma
- UNC Blood Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Tarun Tyagi
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| | - Silvio Antoniak
- UNC Blood Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
18
|
He H, Yang W, Su N, Zhang C, Dai J, Han F, Singhal M, Bai W, Zhu X, Zhu J, Liu Z, Xia W, Liu X, Zhang C, Jiang K, Huang W, Chen D, Wang Z, He X, Kirchhoff F, Li Z, Liu C, Huan J, Wang X, Wei W, Wang J, Augustin HG, Hu J. Activating NO-sGC crosstalk in the mouse vascular niche promotes vascular integrity and mitigates acute lung injury. J Exp Med 2022; 220:213673. [PMID: 36350314 PMCID: PMC9984546 DOI: 10.1084/jem.20211422] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/20/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022] Open
Abstract
Disruption of endothelial cell (ECs) and pericytes interactions results in vascular leakage in acute lung injury (ALI). However, molecular signals mediating EC-pericyte crosstalk have not been systemically investigated, and whether targeting such crosstalk could be adopted to combat ALI remains elusive. Using comparative genome-wide EC-pericyte crosstalk analysis of healthy and LPS-challenged lungs, we discovered that crosstalk between endothelial nitric oxide and pericyte soluble guanylate cyclase (NO-sGC) is impaired in ALI. Indeed, stimulating the NO-sGC pathway promotes vascular integrity and reduces lung edema and inflammation-induced lung injury, while pericyte-specific sGC knockout abolishes this protective effect. Mechanistically, sGC activation suppresses cytoskeleton rearrangement in pericytes through inhibiting VASP-dependent F-actin formation and MRTFA/SRF-dependent de novo synthesis of genes associated with cytoskeleton rearrangement, thereby leading to the stabilization of EC-pericyte interactions. Collectively, our data demonstrate that impaired NO-sGC crosstalk in the vascular niche results in elevated vascular permeability, and pharmacological activation of this crosstalk represents a promising translational therapy for ALI.
Collapse
Affiliation(s)
- Hao He
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Wu Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Nan Su
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Chuankai Zhang
- Department of Burn and Plastic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianing Dai
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Feng Han
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Mahak Singhal
- Laboratory of AngioRhythms, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Wenjuan Bai
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolan Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Jing Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Zhen Liu
- University of Chinese Academy of Sciences, Beijing, China,Chinese Academy of Sciences Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Wencheng Xia
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoting Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Chonghe Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Kai Jiang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Wenhui Huang
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Dan Chen
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zhaoyin Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Xueyang He
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Frank Kirchhoff
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg, Germany
| | - Zhenyu Li
- Texas A&M Health Science Center, Bryan, TX
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China
| | - Jingning Huan
- Department of Burn and Plastic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohong Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Wu Wei
- University of Chinese Academy of Sciences, Beijing, China,Chinese Academy of Sciences Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Jing Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hellmut G. Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Junhao Hu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China,University of Chinese Academy of Sciences, Beijing, China,Correspondence to Junhao Hu:
| |
Collapse
|
19
|
Zhang Y, Wu C, Li L, Pandeya A, Zhang G, Cui J, Kirchhofer D, Wood JP, Smyth SS, Wei Y, Li Z. Extracellular Histones Trigger Disseminated Intravascular Coagulation by Lytic Cell Death. Int J Mol Sci 2022; 23:ijms23126800. [PMID: 35743244 PMCID: PMC9224270 DOI: 10.3390/ijms23126800] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/02/2022] [Accepted: 06/16/2022] [Indexed: 01/08/2023] Open
Abstract
Histones are cationic nuclear proteins that are essential for the structure and functions of eukaryotic chromatin. However, extracellular histones trigger inflammatory responses and contribute to death in sepsis by unknown mechanisms. We recently reported that inflammasome activation and pyroptosis trigger coagulation activation through a tissue-factor (TF)-dependent mechanism. We used a combination of various deficient mice to elucidate the molecular mechanism of histone-induced coagulation. We showed that histones trigger coagulation activation in vivo, as evidenced by coagulation parameters and fibrin deposition in tissues. However, histone-induced coagulopathy was neither dependent on intracellular inflammasome pathways involving caspase 1/11 and gasdermin D (GSDMD), nor on cell surface receptor TLR2- and TLR4-mediated host immune response, as the deficiency of these genes in mice did not protect against histone-induced coagulopathy. The incubation of histones with macrophages induced lytic cell death and phosphatidylserine (PS) exposure, which is required for TF activity, a key initiator of coagulation. The neutralization of TF diminished the histone-induced coagulation. Our findings revealed lytic cell death as a novel mechanism of histone-induced coagulation activation and thrombosis.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China;
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY 40506, USA; (G.Z.); (J.P.W.)
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A & M University, College Station, TX 76549, USA;
| | - Congqing Wu
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY 40506, USA; (G.Z.); (J.P.W.)
- Department of Surgery, College of Medicine, University of Kentucky, Lexington, KY 40506, USA
- Correspondence: (C.W.); (Z.L.)
| | - Lan Li
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY 40506, USA; (L.L.); (A.P.); (J.C.)
| | - Ankit Pandeya
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY 40506, USA; (L.L.); (A.P.); (J.C.)
| | - Guoying Zhang
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY 40506, USA; (G.Z.); (J.P.W.)
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A & M University, College Station, TX 76549, USA;
| | - Jian Cui
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY 40506, USA; (L.L.); (A.P.); (J.C.)
| | - Daniel Kirchhofer
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA 94080, USA;
| | - Jeremy P. Wood
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY 40506, USA; (G.Z.); (J.P.W.)
| | - Susan S. Smyth
- Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Yinan Wei
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A & M University, College Station, TX 76549, USA;
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY 40506, USA; (L.L.); (A.P.); (J.C.)
| | - Zhenyu Li
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY 40506, USA; (G.Z.); (J.P.W.)
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A & M University, College Station, TX 76549, USA;
- Correspondence: (C.W.); (Z.L.)
| |
Collapse
|
20
|
Johnson C, Quach HQ, Lau C, Ekholt K, Espevik T, Woodruff TM, Pischke SE, Mollnes TE, Nilsson PH. Thrombin Differentially Modulates the Acute Inflammatory Response to Escherichia coli and Staphylococcus aureus in Human Whole Blood. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2771-2778. [PMID: 35675954 DOI: 10.4049/jimmunol.2101033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 04/11/2022] [Indexed: 06/15/2023]
Abstract
Thrombin plays a central role in thromboinflammatory responses, but its activity is blocked in the common ex vivo human whole blood models, making an ex vivo study of thrombin effects on thromboinflammatory responses unfeasible. In this study, we exploited the anticoagulant peptide Gly-Pro-Arg-Pro (GPRP) that blocks fibrin polymerization to study the effects of thrombin on acute inflammation in response to Escherichia coli and Staphylococcus aureus Human blood was anticoagulated with either GPRP or the thrombin inhibitor lepirudin and incubated with either E. coli or S. aureus for up to 4 h at 37°C. In GPRP-anticoagulated blood, there were spontaneous elevations in thrombin levels and platelet activation, which further increased in the presence of bacteria. Complement activation and the expression of activation markers on monocytes and granulocytes increased to the same extent in both blood models in response to bacteria. Most cytokines were not elevated in response to thrombin alone, but thrombin presence substantially and heterogeneously modulated several cytokines that increased in response to bacterial incubations. Bacterial-induced releases of IL-8, MIP-1α, and MIP-1β were potentiated in the thrombin-active GPRP model, whereas the levels of IP-10, TNF, IL-6, and IL-1β were elevated in the thrombin-inactive lepirudin model. Complement C5-blockade, combined with CD14 inhibition, reduced the overall cytokine release significantly, both in thrombin-active and thrombin-inactive models. Our data support that thrombin itself marginally induces leukocyte-dependent cytokine release in this isolated human whole blood but is a significant modulator of bacteria-induced inflammation by a differential effect on cytokine patterns.
Collapse
Affiliation(s)
- Christina Johnson
- Department of Immunology, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Huy Quang Quach
- Department of Immunology, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Corinna Lau
- Research Laboratory, Nordland Hospital, Bodø, Norway
| | - Karin Ekholt
- Department of Immunology, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Terje Espevik
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Søren Erik Pischke
- Department of Immunology, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway
- Clinic for Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
| | - Tom Eirik Mollnes
- Department of Immunology, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway
- Research Laboratory, Nordland Hospital, Bodø, Norway
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway; and
| | - Per H Nilsson
- Department of Immunology, University of Oslo and Oslo University Hospital Rikshospitalet, Oslo, Norway;
- Department of Chemistry and Biomedicine, Linnaeus Centre for Biomaterials Chemistry Linnaeus University, Kalmar, Sweden
| |
Collapse
|
21
|
Li Y, Feng G. TLR4 inhibitor alleviates sepsis-induced organ failure by inhibiting platelet mtROS production, autophagy, and GPIIb/IIIa expression. J Bioenerg Biomembr 2022; 54:155-162. [PMID: 35676565 DOI: 10.1007/s10863-022-09940-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/24/2022] [Indexed: 10/18/2022]
Abstract
Thrombocytopenia and impaired platelet function are associated with sepsis-induced organ failure. Numerous studies have shown that mitochondrial ROS (mtROS) and autophagy are related to organ injury in sepsis. However, the relationships between platelet mtROS, autophagy and sepsis organ failure remain unclear. Herein, we explored whether toll like receptor 4 (TLR4) inhibitor alleviates sepsis organ failure by inhibiting platelet mtROS production, autophagy, and GPIIb/IIIa expression.Mice were administrated with LPS, LPS + TAK242 or vehicle. The lungs and kidneys were harvested and analyzed using hematoxylin and eosin staining assay. Platelet rich plasma (PRP) was isolated from blood and platelets aggregation and TLR4 expression were analyzed using flow cytometer and western blot. PRP from healthy volunteers was treated with saline, LPS, or LPS + TAK242, and then mitoSOX and calcium were detected using flow cytometer, and NOX2 and LC3B were tested using western blot.Results showed that TAK242 effectively alleviated LPS-induced acute kidney and lung injury in mice, and decreased CD41 expression more significantly than CD62P. In vitro, by inhibiting TLR4, TAK242 suppressed Ca2+, mitoSOX fluorescence, NOX2 expression and LC3BII/LC3BI ratio in LPS treated platelets.TLR4 inhibitor TAK242 may effectively alleviate mouse lung and kidney injury by inhibition of mouse platelet GPIIb/IIIa, and reduce LPS-induced mtROS generation related to Ca2+ influx, thus reducing platelet activation.
Collapse
Affiliation(s)
- Ying Li
- Department of Hematology, the Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Changsha, Hunan, People's Republic of China
| | - Guo Feng
- Department of Nutrition, the Third Xiangya Hospital, No. 138 Tongzipo Road, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
22
|
Jiang J, Liu D, Wang Y, Li W, Hong Z, An J, Qiao S, Xie Z. Glaucocalyxin a protect liver function via inhibiting platelet over-activation during sepsis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 100:154089. [PMID: 35398736 DOI: 10.1016/j.phymed.2022.154089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Rabdosia japonica (Burm. f.) var. glaucocalyx (Maxim.) is a perennial herb, and is traditionally used as folk medicine for treating inflammatory diseases and cancer. Gaucocalyxin A (GLA) is an ent‑kaurane diterpenoid that is isolated from the aerial parts of R. japonica (Burm. f.) var. glaucocalyx (Maxim.). In a recent study, we found that GLA protects against acute liver dysfunction induced by Escherichia coli, which is likely related to its anti-inflammatory effects. However, the mechanism by which GLA protects liver injury during sepsis is unknown. AIM To evaluate the anti-inflammatory function of GLA and its regulatory effect on platelet function. METHOD An in vivo model of sepsis was established by inoculating mice with E. coli. Live function and platelet activation were evaluated through standard assays. The levels of pro-inflammatory factors were measured through ELISA and qRT-PCR. RESULTS GLA alleviated liver dysfunction in the mouse model of sepsis. GLA-treated mice displayed lower complement activation and liver dysfunction after E. coli infection. GLA alleviated the decrease in peripheral platelet counts by inhibiting their clearance by Kupffer cells in liver. Furthermore, GLA inhibited platelet activation through the RIP1/RIP3/AKT pathway and downregulated C3aR expression on the platelets, thereby inhibiting liver injury and dysfunction due to excessive complement activation. CONCLUSION GLA can inhibit platelet activation by reducing surface expression of C3aR, which protect the liver from injury induced by excessive complement activation. GLA is a novel therapeutic agent for controlling sepsis-related liver dysfunction.
Collapse
Affiliation(s)
- Jiang Jiang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Dengping Liu
- Institute of Clinical Medicine Research, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Yuanyuan Wang
- Department of Intensive Care Unit, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Wei Li
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Zhihui Hong
- Department of Nuclear Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianzhong An
- Institute of Clinical Medicine Research, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Shigang Qiao
- Institute of Clinical Medicine Research, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, China; Faculty of Anesthesiology, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Zhanli Xie
- Institute of Clinical Medicine Research, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, China.
| |
Collapse
|
23
|
Davenport P, Fan HH, Nolton E, Feldman HA, Lorenz V, Canas J, Acosta-Zaldívar M, Yakah W, Arthur C, Martin C, Stowell S, Koehler J, Mager D, Sola-Visner M. Platelet transfusions in a murine model of neonatal polymicrobial sepsis: Divergent effects on inflammation and mortality. Transfusion 2022; 62:1177-1187. [PMID: 35522536 PMCID: PMC11465244 DOI: 10.1111/trf.16895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/11/2022] [Accepted: 03/18/2022] [Indexed: 12/19/2022]
Abstract
BACKGROUND Platelet transfusions (PTxs) are often given to septic preterm neonates at high platelet count thresholds in an attempt to reduce bleeding risk. However, the largest randomized controlled trial (RCT) of neonatal transfusion thresholds found higher mortality and/or major bleeding in infants transfused at higher thresholds. Using a murine model, we investigated the effects of adult PTx on neonatal sepsis-induced mortality, systemic inflammation, and platelet consumption. STUDY DESIGN AND METHODS Polymicrobial sepsis was induced via intraperitoneal injection of cecal slurry preparations (CS1, 2, 3) into P10 pups. Two hours after infection, pups were transfused with washed adult Green Flourescent Protein (GFP+) platelets or control. Weights, platelet counts, and GFP% were measured before 4 and 24 h post-infection. At 24 h, blood was collected for quantification of plasma cytokines. RESULTS The CS batches varied in 24 h mortality (11%, 73%, and 30% in CS1, 2, and 3, respectively), due to differences in bacterial composition. PTx had differential effects on sepsis-induced mortality and systemic inflammatory cytokines, increasing both in mice infected with CS1 (low mortality) and decreasing both in mice infected with CS2 and 3. In a mathematical model of platelet kinetics, the consumption of transfused adult platelets was higher than that of endogenous neonatal platelets, regardless of CS batch. DISCUSSION Our findings support the hypothesis that transfused adult platelets are consumed faster than endogenous neonatal platelets in sepsis and demonstrate that PTx can enhance or attenuate neonatal inflammation and mortality in a model of murine polymicrobial sepsis, depending on the composition of the inoculum and/or the severity of sepsis.
Collapse
Affiliation(s)
- Patricia Davenport
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Hsuan-Hao Fan
- Department of Pharmaceutical Sciences, University of Buffalo, State University of New York, Buffalo, NY
| | - Emily Nolton
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA
| | - Henry A. Feldman
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Institutional Centers for Clinical and Translational Research, Boston Children’s Hospital, Boston, MA
| | - Viola Lorenz
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Jorge Canas
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA
| | | | - William Yakah
- Harvard Medical School, Boston, MA
- Division of Neonatology, Beth Israel Medical Center, Boston, MA
| | - Connie Arthur
- Harvard Medical School, Boston, MA
- Transfusion Medicine, Brigham and Women Hospital, Boston, MA
| | - Camilia Martin
- Harvard Medical School, Boston, MA
- Division of Neonatology, Beth Israel Medical Center, Boston, MA
| | - Sean Stowell
- Harvard Medical School, Boston, MA
- Transfusion Medicine, Brigham and Women Hospital, Boston, MA
| | - Julia Koehler
- Harvard Medical School, Boston, MA
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA
| | - Donald Mager
- Department of Pharmaceutical Sciences, University of Buffalo, State University of New York, Buffalo, NY
| | - Martha Sola-Visner
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
24
|
Liu X, Bao Y, Lin Z, Tang L, Mao P. Platelets inhibit development of atherosclerosis in atherosclerotic mice. Cell Cycle 2022; 21:1222-1232. [PMID: 35213268 PMCID: PMC9103360 DOI: 10.1080/15384101.2022.2044703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Platelets can protect from lipopolysaccharide-induced septic shock by inhibiting inflammation, but it is unknown whether platelets have an anti-atherosclerotic effect. The aim of this study was to investigate the effect of platelet transfusion on atherosclerosis (AS) in a mouse model of AS. Apolipoprotein E deficiency (ApoE-/-) mice were fed with a high-fat diet (HFD) for 8 weeks to establish a mouse model of AS. Mice weekly underwent bi-weekly injection with or without platelets during AS induction (HFD+platelet). Hematoxylin-eosin (H&E), Oil Red O, and Sudan IV stainings were used to assess pathological and morphological changes in the aortic tissue. Lipid levels, and liver and kidney function were examined using an automatic biochemical analyzer. Immune histochemical assays were used to detect the infiltration and distribution of inflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin 1β (IL-1β), IL-6, and monocyte chemotactic protein-1 (MCP-1) in the aortic arch. Western blot and enzyme-linked immunosorbent assay (ELISA) were used to examine the expression levels of TNF-α, IL-1β, IL-6, and MCP-1 in the aorta or the peripheral blood, respectively. Compared with the HFD group, AS pathological lesions from the aortic arch in the HFD+platelet group were significantly smaller and alterations in the lipid metabolism were also less pronounced. Furthermore, TNF-α, IL-1β, IL-6, and MCP-1 levels were all significantly reduced in mice that received platelet injection. Platelets transfusion can effectively ameliorate lipid metabolism, suppress the inflammatory response in the vascular wall, and inhibit the development of AS in mice.
Collapse
Affiliation(s)
- Xiaowei Liu
- Department of Cardiology, Zhejiang Hospital, Hangzhou, P. R. China
| | - Yizhong Bao
- Zhejiang Provincial Key Laboratory of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, P. R. China
| | - Zhang Lin
- Department of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, P. R. China
| | - Lijiang Tang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, P. R. China,Lijiang Tang Department of Cardiology, Zhejiang Hospital, 12 Lingying Road, Hangzhou, Zhejiang 310013, P. R. China Department of Cardiology, Zhejiang Hospital, Hangzhou, P. R. China
| | - Ping Mao
- Department of Cardiology, Zhejiang Hospital, Hangzhou, P. R. China,CONTACT Ping Mao Department of Cardiology, Zhejiang Hospital, 12 Lingying Road, Hangzhou, Zhejiang310013, P. R. China
| |
Collapse
|
25
|
Mas-Bargues C, Borrás C, Viña J. The multimodal action of genistein in Alzheimer's and other age-related diseases. Free Radic Biol Med 2022; 183:127-137. [PMID: 35346775 DOI: 10.1016/j.freeradbiomed.2022.03.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/07/2023]
Abstract
Genistein is a phytoestrogen that, due to its structural similarity with estrogen, can both mimic and antagonize estrogen effects. Early analysis proved that at high concentrations, genistein inhibits breast cancer cell proliferation, thereby suggesting an anticancer activity. Since then, many discoveries have identified the genistein mechanism of action, including cell cycle arrest, apoptosis induction, as well as angiogenesis, and metastasis inhibition. In this review, we aim to discuss the multimodal action of genistein as an antioxidant, anti-inflammatory, anti-amyloid β, and autophagy promoter, which could be responsible for the genistein beneficial effect on Alzheimer's. Furthermore, we pinpoint the main signal transduction pathways that are known to be modulated by genistein. Genistein has thus several beneficial effects in several diseases, many of them associated with age, such as the above mentioned Alzheimer disease. Indeed, the beneficial effects of genistein for health promotion depend on each multimodality. In the context of geroscience, genistein has promising beneficial effects due to its multimodal action to treat age associated-diseases.
Collapse
Affiliation(s)
- Cristina Mas-Bargues
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, Valencia, 46010, Spain.
| | - Consuelo Borrás
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, Valencia, 46010, Spain.
| | - José Viña
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, Valencia, 46010, Spain
| |
Collapse
|
26
|
Valet C, Magnen M, Qiu L, Cleary SJ, Wang KM, Ranucci S, Grockowiak E, Boudra R, Conrad C, Seo Y, Calabrese DR, Greenland JR, Leavitt AD, Passegué E, Méndez-Ferrer S, Swirski FK, Looney MR. Sepsis promotes splenic production of a protective platelet pool with high CD40 ligand expression. J Clin Invest 2022; 132:e153920. [PMID: 35192546 PMCID: PMC8970674 DOI: 10.1172/jci153920] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 02/16/2022] [Indexed: 11/17/2022] Open
Abstract
Platelets have a wide range of functions including critical roles in hemostasis, thrombosis, and immunity. We hypothesized that during acute inflammation, such as in life-threatening sepsis, there are fundamental changes in the sites of platelet production and phenotypes of resultant platelets. Here, we showed during sepsis that the spleen was a major site of megakaryopoiesis and platelet production. Sepsis provoked an adrenergic-dependent mobilization of megakaryocyte-erythrocyte progenitors (MEPs) from the bone marrow to the spleen, where IL-3 induced their differentiation into megakaryocytes (MKs). In the spleen, immune-skewed MKs produced a CD40 ligandhi platelet population with potent immunomodulatory functions. Transfusions of post-sepsis platelets enriched from splenic production enhanced immune responses and reduced overall mortality in sepsis-challenged animals. These findings identify a spleen-derived protective platelet population that may be broadly immunomodulatory in acute inflammatory states such as sepsis.
Collapse
Affiliation(s)
- Colin Valet
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Mélia Magnen
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Longhui Qiu
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Simon J. Cleary
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Kristin M. Wang
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Serena Ranucci
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Elodie Grockowiak
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Rafik Boudra
- Department of Dermatology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Yurim Seo
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | | | | | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York, USA
| | - Simón Méndez-Ferrer
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge, United Kingdom
| | - Filip K. Swirski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Mark R. Looney
- Department of Medicine, UCSF, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| |
Collapse
|
27
|
Mandel J, Casari M, Stepanyan M, Martyanov A, Deppermann C. Beyond Hemostasis: Platelet Innate Immune Interactions and Thromboinflammation. Int J Mol Sci 2022; 23:ijms23073868. [PMID: 35409226 PMCID: PMC8998935 DOI: 10.3390/ijms23073868] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 02/07/2023] Open
Abstract
There is accumulating evidence that platelets play roles beyond their traditional functions in thrombosis and hemostasis, e.g., in inflammatory processes, infection and cancer, and that they interact, stimulate and regulate cells of the innate immune system such as neutrophils, monocytes and macrophages. In this review, we will focus on platelet activation in hemostatic and inflammatory processes, as well as platelet interactions with neutrophils and monocytes/macrophages. We take a closer look at the contributions of major platelet receptors GPIb, αIIbβ3, TLT-1, CLEC-2 and Toll-like receptors (TLRs) as well as secretions from platelet granules on platelet-neutrophil aggregate and neutrophil extracellular trap (NET) formation in atherosclerosis, transfusion-related acute lung injury (TRALI) and COVID-19. Further, we will address platelet-monocyte and macrophage interactions during cancer metastasis, infection, sepsis and platelet clearance.
Collapse
Affiliation(s)
- Jonathan Mandel
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; (J.M.); (M.C.); (M.S.)
| | - Martina Casari
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; (J.M.); (M.C.); (M.S.)
| | - Maria Stepanyan
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; (J.M.); (M.C.); (M.S.)
- Center For Theoretical Problems of Physico-Chemical Pharmacology, 109029 Moscow, Russia;
- Physics Faculty, Lomonosov Moscow State University, 119991 Moscow, Russia
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology Immunology Ministry of Healthcare of Russian Federation, 117198 Moscow, Russia
| | - Alexey Martyanov
- Center For Theoretical Problems of Physico-Chemical Pharmacology, 109029 Moscow, Russia;
- Dmitriy Rogachev National Medical Research Center of Pediatric Hematology, Oncology Immunology Ministry of Healthcare of Russian Federation, 117198 Moscow, Russia
- N.M. Emanuel Institute of Biochemical Physics RAS (IBCP RAS), 119334 Moscow, Russia
| | - Carsten Deppermann
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; (J.M.); (M.C.); (M.S.)
- Correspondence:
| |
Collapse
|
28
|
Wang R, Dai H. Association of platelet count with all-cause mortality from acute respiratory distress syndrome: A cohort study. J Clin Lab Anal 2022; 36:e24378. [PMID: 35358347 PMCID: PMC9102613 DOI: 10.1002/jcla.24378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background The purpose of this study was to investigate whether platelet count was associated with mortality in acute respiratory distress syndrome (ARDS) patients. Methods We analyzed patients with ARDS from Multi‐parameter Intelligent Monitoring in Intensive Care Database III (MIMIC‐III). Platelet count was measured at the time of intensive care unit (ICU) admission. The cox proportional hazard model and subgroup analysis were used to determine the relationship between the platelet count and mortality of ARDS, as well as the consistency of its association. The primary outcome of this study was 365‐day mortality from the date of ICU admission. Result This study enrolled a total of 395 critically ill patients with ARDS. After adjustment for age, gender and ethnicity, the multivariate cox regression model showed that the hazard ratios (HRs) (95% confidence intervals [CIs]) of platelet count <192 × 109/L and >296 × 109/L were 2.08 (1.43, 3.04) and 1.35 (0.91, 2.01), respectively, compared with the reference (192–296 ×109/L). After adjusting for confounding factors, lower platelet count (<192 × 109/L) was associated with increased mortality (adjusted HR, 1.71; 95% CI 1.06–2.76, p = 0.0284). However, there was no similar trend in the 30‐day (adjusted HR,1.02; 95% CI 0.54–1.94) or 90‐day (adjusted HR, 1.65; 95% CI 0.94–2.89) mortality. In the subgroup analysis, lower platelet count showed significant interactions with specific populations (p interaction = 0.0413), especially in patients with atrial fibrillation. Conclusion Taken together, our analysis showed that platelet count is an independent predictor of mortality in critically ill patients with ARDS.
Collapse
Affiliation(s)
- Rennv Wang
- Emergency Department, Affiliated Zhejiang Hospital of Zhejiang University School of Medical, Hangzhou, Zhejiang, China
| | - Haiwen Dai
- Emergency Department, Affiliated Zhejiang Hospital of Zhejiang University School of Medical, Hangzhou, Zhejiang, China
| |
Collapse
|
29
|
Nociceptor-derived Reg3γ prevents endotoxic death by targeting kynurenine pathway in microglia. Cell Rep 2022; 38:110462. [PMID: 35263589 DOI: 10.1016/j.celrep.2022.110462] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 01/11/2022] [Accepted: 02/09/2022] [Indexed: 11/21/2022] Open
Abstract
Nociceptors can fine-tune local or systemic immunity, but the mechanisms of nociceptive modulation in endotoxic death remain largely unknown. Here, we identified C-type lectin Reg3γ as a nociceptor-enriched hormone that protects the host from endotoxic death. During endotoxemia, nociceptor-derived Reg3γ penetrates the brain and suppresses the expression of microglial indoleamine dioxygenase 1, a critical enzyme of the kynurenine pathway, via the Extl3-Bcl10 axis. Endotoxin-administered nociceptor-null mice and nociceptor-specific Reg3γ-deficient mice exhibit a high mortality rate accompanied by decreased brain HK1 phosphorylation and ATP production despite normal peripheral inflammation. Such metabolic arrest is only observed in the brain, and aberrant production of brain quinolinic acid, a neurotoxic metabolite of the kynurenine pathway, causes HK1 suppression. Strikingly, the central administration of Reg3γ protects mice from endotoxic death by enhancing brain ATP production. By identifying nociceptor-derived Reg3γ as a microglia-targeted hormone, this study provides insights into the understanding of tolerance to endotoxic death.
Collapse
|
30
|
Rabouël Y, Magnenat S, Lefebvre F, Delabranche X, Gachet C, Hechler B. Transfusion of fresh washed platelets does not prevent experimental polymicrobial-induced septic shock in mice. J Thromb Haemost 2022; 20:449-460. [PMID: 34752015 DOI: 10.1111/jth.15583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 11/29/2022]
Abstract
INTRODUCTION The specific role of platelets during sepsis is not yet fully understood, probably related to the paradox of platelets being potentially beneficial but also deleterious via their thrombotic functions. OBJECTIVE To evaluate the impact of thrombocytopenia on septic shock in mice and to investigate whether transfusion of fresh washed platelets, either fully functional or with impaired hemostatic properties, might have beneficial effects. METHODS Septic shock was induced by cecal ligation and puncture (CLP). Experimental depletion of circulating platelets was induced with a rat anti-mouse GPIbα monoclonal antibody. Transfusion of either wild-type washed platelets, platelets treated with the antiplatelet drugs acetylsalicylic acid (ASA) and clopidogrel, or GPIIbIIIa-deficient washed platelets treated with ASA and clopidogrel was performed 4 h after CLP surgery. RESULTS Depletion of circulating platelets negatively affected septic shock, worsening systemic inflammation, coagulopathy, organ damage, and mortality, raising the question of whether a higher platelet count could be protective. Transfusion of fully functional platelets or platelets with combined treatment with ASA and clopidogrel, with or without additional GPIIbIIIa deficiency, afforded an immediate return of circulating platelet counts to their initial values before surgery. However, transfusion of each of the three types of platelets did not prevent arterial hypotension, inflammatory response, coagulopathy, and organ damage during septic shock. CONCLUSION Depletion of circulating platelets negatively affects septic shock, while transfusion of washed platelets has no significant beneficial effect in mice.
Collapse
Affiliation(s)
- Yannick Rabouël
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand Est, BPPS UMR_S1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Stéphanie Magnenat
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand Est, BPPS UMR_S1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Floryna Lefebvre
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand Est, BPPS UMR_S1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Xavier Delabranche
- Hôpitaux Universitaires de Strasbourg, Anesthésie, Réanimation et Médecine périopératoire, Nouvel Hôpital Civil, Strasbourg, France
| | - Christian Gachet
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand Est, BPPS UMR_S1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Beatrice Hechler
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand Est, BPPS UMR_S1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| |
Collapse
|
31
|
Liu X, Chen X, Xu C, Lou J, Weng Y, Tang L. Platelet protects angiotensin II-driven abdominal aortic aneurysm formation through inhibition of inflammation. Exp Gerontol 2022; 159:111703. [PMID: 35038567 DOI: 10.1016/j.exger.2022.111703] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 01/16/2023]
Abstract
BACKGROUND Inflammation is the primary cause of abdominal aortic aneurysm (AAA) formation and development. It has been reported that platelets protect against septic shock by inhibiting inflammation. However, it is unclear whether platelets protect AAA progress via suppressing inflammation. METHODS A mouse model of AAA was established by a daily administration of angiotensin II (Ang II, 1000 ng/kg/min) for 28-day. The AAA mice received 1 × 109 platelets transfusion in normal saline every 3rd day for 1 month. Hematoxylin and eosin, Masson's trichrome, and elastic van Gieson staining techniques were used to analyze the morphology of the abdominal aorta. Immunohistochemistry was used to detect any infiltration of inflammatory cells, inflammatory factors, and matrix metalloproteins (MMPs) in the aortic tissue. Western blot and enzyme-linked immunosorbent assay (ELISA) were used to examine the inflammatory factor proteins levels in the aortic wall and peripheral blood, respectively. RESULTS Platelets infusion significantly suppressed the Ang II-driven elevation of aortic diameter, AAA formation (52.38% decrease, P < 0.05), aortic expansion, elastic lamina destruction, and inflammatory response. In addition, MMP-2 and MMP-9 production were also reduced by platelets transfusion. CONCLUSIONS For the first time, our study reported the beneficial effect of platelet transfusion in suppressing the Ang II-driven AAA progress in mice through the inhibition of inflammation.
Collapse
Affiliation(s)
- Xiaowei Liu
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, PR China
| | - Xiaofeng Chen
- Department of Cardiology, Taizhou Hospital, Linhai, Zhejiang 317000, PR China
| | - Chen Xu
- Department of Cardiology, Taizhou Hospital, Linhai, Zhejiang 317000, PR China
| | - Jiangjie Lou
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, PR China
| | - Yingzheng Weng
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, PR China
| | - Lijiang Tang
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, PR China.
| |
Collapse
|
32
|
Zheng L, Duan Z, Tang D, He Y, Chen X, Chen Q, Li M. GP IIb/IIIa-Mediated Platelet Activation and Its Modulation of the Immune Response of Monocytes Against Candida albicans. Front Cell Infect Microbiol 2021; 11:783085. [PMID: 34938671 PMCID: PMC8685400 DOI: 10.3389/fcimb.2021.783085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Candida albicans is the most common fungal pathogen in humans, causing invasive disease and even potentially life-threatening systemic infections when tissue homeostasis is disrupted. Previous studies have identified an essential role of platelets in infection and immunity, especially when they are activated. However, it is still unclear whether platelets can be activated by C. albicans, and even less is known about the role of platelets in C. albicans infection. Herein, we showed that C. albicans induced platelet activation in vitro. C. albicans elevated the levels of AKT Ser473 phosphorylation, and inhibition of the PI3K-AKT signaling pathway reversed C. albicans-induced platelet activation. Surprisingly, C. albicans-induced platelet activation occurred in an integrin glycoprotein (GP) IIb/IIIa-dependent manner but was independent of the pattern recognition receptors toll-like receptor (TLR) 2 and TLR4. Interestingly, platelets enhanced the phagocytosis of human monocytes challenged with C. albicans and upregulated the expression of inflammatory cytokines, which were dependent on platelet activation mediated by GP IIb/IIIa. The present work provides new insights into the role of activated platelets in the defense against C. albicans, highlighting the importance of GP IIb/IIIa in the recognition of C. albicans.
Collapse
Affiliation(s)
- Lin Zheng
- Institute of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and Sexually Transmitted Infections, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Zhimin Duan
- Institute of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and Sexually Transmitted Infections, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | | | - Yanzhi He
- School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xu Chen
- Institute of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and Sexually Transmitted Infections, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.,School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qing Chen
- Jiangsu Province Blood Center, Nanjing, China.,Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Min Li
- Institute of Dermatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and Sexually Transmitted Infections, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.,Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
33
|
Tokarz-Deptuła B, Palma J, Baraniecki Ł, Stosik M, Kołacz R, Deptuła W. What Function Do Platelets Play in Inflammation and Bacterial and Viral Infections? Front Immunol 2021; 12:770436. [PMID: 34970260 PMCID: PMC8713818 DOI: 10.3389/fimmu.2021.770436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/22/2021] [Indexed: 12/15/2022] Open
Abstract
The article presents the function of platelets in inflammation as well as in bacterial and viral infections, which are the result of their reaction with the endovascular environment, including cells of damaged vascular endothelium and cells of the immune system. This role of platelets is conditioned by biologically active substances present in their granules and in their specific structures - EV (extracellular vesicles).
Collapse
Affiliation(s)
| | - Joanna Palma
- Department of Biochemical Sciences, Pomeranian Medical University, Szczecin, Poland
| | | | - Michał Stosik
- Institute of Biological Science, Faculty of Biological Sciences, University of Zielona Góra, Zielona Góra, Poland
| | - Roman Kołacz
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Wiesław Deptuła
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Toruń, Poland
| |
Collapse
|
34
|
Yang JL, Yang CJ, Chuang YC, Sheng WH, Chen YC, Chang SC. Association of capsular polysaccharide locus 2 with prognosis of Acinetobacter baumannii bacteraemia. Emerg Microbes Infect 2021; 11:83-90. [PMID: 34825848 PMCID: PMC8725928 DOI: 10.1080/22221751.2021.2011624] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Acinetobacter baumannii causes healthcare-associated infections worldwide. Capsular polysaccharide (CPS) is shown an important virulence factor of A. baumannii both in vitro and in vivo. Capsule locus 2 (KL2) for CPS is the most common KL type and is associated with carbapenem resistance. It is unclear whether KL2 is related to the clinical outcome of invasive A. baumannii infection. Here we had followed patients with A. baumannii bacteraemia prospectively between 2009 and 2014. One-third of the unduplicated blood isolates were randomly selected each year for microbiological and clinical studies. The KL2 gene cluster was identified using polymerase chain reaction. A total of 148 patients were enrolled randomly. Eighteen isolates (12.2%) carried KL2, and 130 isolates (87.8%) didn’t. Compared with non-KL2 isolates, KL2 isolates had significantly higher resistance to imipenem, sulbactam, and tigecycline. Compared with the non-KL group, in the KL2 group, the hospital stay before development of bacteraemia was longer (P < 0.001), a higher percentage had pneumonia (P = 0.004), and the white blood cell count was lower (P = 0.03). Infection with KL2 A. baumannii predicted mortality (adjusted hazard ratio [aHR], 2.03; 95% confidence interval [CI], 1.09–3.78; P = 0.03), independently of the Pitt bacteraemia score (aHR, 1.34; 95% CI, 1.23–1.46; P < 0.001) and leucopenia (aHR, 2.16; 95% CI, 1.30–3.57; P = 0.003). Thrombocytopenia contributed to the effect of KL2 on mortality in bacteraemia (Sobel test P = 0.01). Large-scale studies are warranted to confirm these findings and the underlying mechanisms deserve further investigation.
Collapse
Affiliation(s)
- Jia-Ling Yang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Jui Yang
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chung Chuang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wang-Huei Sheng
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yee-Chun Chen
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shan-Chwen Chang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
35
|
Lin D, Shen L, Luo M, Zhang K, Li J, Yang Q, Zhu F, Zhou D, Zheng S, Chen Y, Zhou J. Circulating tumor cells: biology and clinical significance. Signal Transduct Target Ther 2021; 6:404. [PMID: 34803167 PMCID: PMC8606574 DOI: 10.1038/s41392-021-00817-8] [Citation(s) in RCA: 342] [Impact Index Per Article: 114.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/06/2021] [Accepted: 10/27/2021] [Indexed: 02/07/2023] Open
Abstract
Circulating tumor cells (CTCs) are tumor cells that have sloughed off the primary tumor and extravasate into and circulate in the blood. Understanding of the metastatic cascade of CTCs has tremendous potential for the identification of targets against cancer metastasis. Detecting these very rare CTCs among the massive blood cells is challenging. However, emerging technologies for CTCs detection have profoundly contributed to deepening investigation into the biology of CTCs and have facilitated their clinical application. Current technologies for the detection of CTCs are summarized herein, together with their advantages and disadvantages. The detection of CTCs is usually dependent on molecular markers, with the epithelial cell adhesion molecule being the most widely used, although molecular markers vary between different types of cancer. Properties associated with epithelial-to-mesenchymal transition and stemness have been identified in CTCs, indicating their increased metastatic capacity. Only a small proportion of CTCs can survive and eventually initiate metastases, suggesting that an interaction and modulation between CTCs and the hostile blood microenvironment is essential for CTC metastasis. Single-cell sequencing of CTCs has been extensively investigated, and has enabled researchers to reveal the genome and transcriptome of CTCs. Herein, we also review the clinical applications of CTCs, especially for monitoring response to cancer treatment and in evaluating prognosis. Hence, CTCs have and will continue to contribute to providing significant insights into metastatic processes and will open new avenues for useful clinical applications.
Collapse
Affiliation(s)
- Danfeng Lin
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lesang Shen
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meng Luo
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kun Zhang
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinfan Li
- Department of Pathology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Yang
- Department of Pathology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fangfang Zhu
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Zhou
- Department of Surgery, Traditional Chinese Medical Hospital of Zhuji, Shaoxing, China
| | - Shu Zheng
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiding Chen
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jiaojiao Zhou
- Department of Breast Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
36
|
Jiang Y, Tang Y, Hoover C, Kondo Y, Huang D, Restagno D, Shao B, Gao L, Michael McDaniel J, Zhou M, Silasi-Mansat R, McGee S, Jiang M, Bai X, Lupu F, Ruan C, Marth JD, Wu D, Han Y, Xia L. Kupffer cell receptor CLEC4F is important for the destruction of desialylated platelets in mice. Cell Death Differ 2021; 28:3009-3021. [PMID: 33993195 PMCID: PMC8564511 DOI: 10.1038/s41418-021-00797-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/21/2021] [Accepted: 04/26/2021] [Indexed: 02/04/2023] Open
Abstract
The liver has recently been identified as a major organ for destruction of desialylated platelets. However, the underlying mechanism remains unclear. Kupffer cells, which are professional phagocytic cells in the liver, comprise the largest population of resident tissue macrophages in the body. Kupffer cells express a C-type lectin receptor, CLEC4F, that recognizes desialylated glycans with an unclear in vivo role in mediating platelet destruction. In this study, we generated a CLEC4F-deficient mouse model (Clec4f-/-) and found that CLEC4F was specifically expressed by Kupffer cells. Using the Clec4f-/- mice and a newly generated platelet-specific reporter mouse line, we revealed a critical role for CLEC4F on Kupffer cells in mediating destruction of desialylated platelets in the liver in vivo. Platelet clearance experiments and ultrastructural analysis revealed that desialylated platelets were phagocytized predominantly by Kupffer cells in a CLEC4F-dependent manner in mice. Collectively, these findings identify CLEC4F as a Kupffer cell receptor important for the destruction of desialylated platelets induced by bacteria-derived neuraminidases, which provide new insights into the pathogenesis of thrombocytopenia in disease conditions such as sepsis.
Collapse
Affiliation(s)
- Yizhi Jiang
- grid.429222.d0000 0004 1798 0228Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, 215006 China ,grid.452929.10000 0004 8513 0241Department of Hematology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001 China ,grid.274264.10000 0000 8527 6890Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104 USA ,grid.263761.70000 0001 0198 0694Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006 China
| | - Yaqiong Tang
- grid.429222.d0000 0004 1798 0228Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, 215006 China ,grid.274264.10000 0000 8527 6890Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104 USA ,grid.263761.70000 0001 0198 0694Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006 China
| | - Christopher Hoover
- grid.274264.10000 0000 8527 6890Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104 USA
| | - Yuji Kondo
- grid.274264.10000 0000 8527 6890Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104 USA
| | - Dongping Huang
- grid.452929.10000 0004 8513 0241Department of Hematology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001 China
| | - Damien Restagno
- grid.263761.70000 0001 0198 0694State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123 China
| | - Bojing Shao
- grid.274264.10000 0000 8527 6890Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104 USA
| | - Liang Gao
- grid.274264.10000 0000 8527 6890Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104 USA
| | - J. Michael McDaniel
- grid.274264.10000 0000 8527 6890Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104 USA
| | - Meixiang Zhou
- grid.274264.10000 0000 8527 6890Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104 USA
| | - Robert Silasi-Mansat
- grid.274264.10000 0000 8527 6890Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104 USA
| | - Samuel McGee
- grid.274264.10000 0000 8527 6890Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104 USA
| | - Miao Jiang
- grid.429222.d0000 0004 1798 0228Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, 215006 China ,grid.263761.70000 0001 0198 0694Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006 China
| | - Xia Bai
- grid.429222.d0000 0004 1798 0228Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, 215006 China ,grid.263761.70000 0001 0198 0694Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006 China ,grid.263761.70000 0001 0198 0694State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123 China
| | - Florea Lupu
- grid.274264.10000 0000 8527 6890Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104 USA
| | - Changgeng Ruan
- grid.429222.d0000 0004 1798 0228Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, 215006 China ,grid.263761.70000 0001 0198 0694Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006 China ,grid.263761.70000 0001 0198 0694State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123 China
| | - Jamey D. Marth
- grid.133342.40000 0004 1936 9676Center for Nanomedicine, SBP Medical Discovery Institute, and Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, CA 93106 USA
| | - Depei Wu
- grid.429222.d0000 0004 1798 0228Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, 215006 China ,grid.263761.70000 0001 0198 0694Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006 China
| | - Yue Han
- grid.429222.d0000 0004 1798 0228Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, 215006 China ,grid.263761.70000 0001 0198 0694Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006 China
| | - Lijun Xia
- grid.429222.d0000 0004 1798 0228Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, NHC Key Laboratory of Thrombosis and Hemostasis, The First Affiliated Hospital of Soochow University, Suzhou, 215006 China ,grid.274264.10000 0000 8527 6890Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104 USA ,grid.263761.70000 0001 0198 0694Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215006 China
| |
Collapse
|
37
|
Chebbo M, Duez C, Alessi MC, Chanez P, Gras D. Platelets: a potential role in chronic respiratory diseases? Eur Respir Rev 2021; 30:30/161/210062. [PMID: 34526315 PMCID: PMC9488457 DOI: 10.1183/16000617.0062-2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/05/2021] [Indexed: 12/21/2022] Open
Abstract
Platelets are small anucleate cells known for their role in haemostasis and thrombosis. In recent years, an increasing number of observations have suggested that platelets are also immune cells and key modulators of immunity. They express different receptors and molecules that allow them to respond to pathogens, and to interact with other immune cells. Platelets were linked to the pathogenesis of some inflammatory disorders including respiratory diseases such as asthma and idiopathic pulmonary fibrosis. Here, we discuss the involvement of platelets in different immune responses, and we focus on their potential role in various chronic lung diseases. In addition to their essential role in haemostasis and thrombosis, platelets are strong modulators of different immune responses, and could be involved in the physiopathology of several chronic airway diseaseshttps://bit.ly/3cB6Xnj
Collapse
Affiliation(s)
| | | | - Marie C Alessi
- Aix-Marseille Univ, INSERM, INRAE, Marseille, France.,APHM, CHU de la Timone, Laboratoire d'hématologie, Marseille, France
| | - Pascal Chanez
- Aix-Marseille Univ, INSERM, INRAE, Marseille, France.,APHM, Hôpital NORD, Clinique des Bronches, Allergie et Sommeil, Marseille, France
| | - Delphine Gras
- Aix-Marseille Univ, INSERM, INRAE, Marseille, France
| |
Collapse
|
38
|
Makatsariya AD, Slukhanchuk EV, Bitsadze VO, Khizroeva JK, Tretyakova MV, Shkoda AS, Elalamy I, Di Renzo GC, Rizzo G, Pyatigorskaya NV, Solopova AG, Grigoreva KN, Nakaidze IA, Mitryuk DV. The Effect of Various Types of Anticoagulant Therapy on the Reduction of Mortality in COVID-19. ANNALS OF THE RUSSIAN ACADEMY OF MEDICAL SCIENCES 2021. [DOI: 10.15690/vramn1551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a viral infection that, in severe course, leads to the development of a cytokine storm, systemic inflammatory response and coagulopathy. Unlike other sepsis-associated disseminated intravascular coagulopathy, COVID-19 induced coagulopathy is realized mainly in thrombosis. Researchers around the world are currently developing adequate diagnostic, monitoring and anticoagulant therapy approaches to safely and effectively manage patients with severe COVID-19. The need to develop laboratory monitoring is due to the fact that 20% of patients have changes in hemostasis indicators, while in patients with a severe form of the disease, they are present in 100% of cases. In case of deaths from COVID-19, there is an increase in the concentration of D-dimer and fibrinogen degradation products. Thus, the severity of hemostasis disorders has an important prognostic value. Anticoagulant therapy is included in the list of all recommendations as an effective means of reducing mortality from COVID-19. The questions of the recommended groups and doses of anticoagulant drugs are still open. The approach to the choice of an anticoagulant should be based not only on risk factors, characteristics of the course of the disease, anamnesis, but also on the wishes of the patient during long-term therapy at the post-hospital stage.
Collapse
|
39
|
Rabouel Y, Magnenat S, Delabranche X, Gachet C, Hechler B. Platelet P2Y 12 Receptor Deletion or Pharmacological Inhibition does not Protect Mice from Sepsis or Septic Shock. TH OPEN 2021; 5:e343-e352. [PMID: 34447900 PMCID: PMC8384481 DOI: 10.1055/s-0041-1733857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 07/06/2021] [Indexed: 12/27/2022] Open
Abstract
Introduction
Platelets are increasingly appreciated as key effectors during sepsis, raising the question of the usefulness of antiplatelet drugs to treat patients with sepsis.
Objective
Evaluate the potential contribution of the platelet P2Y
12
receptor in the pathogenesis of polymicrobial-induced sepsis and septic shock in mice.
Methods
The effects of P2Y
12
inhibition using clopidogrel treatment and of platelet-specific deletion of the P2Y
12
receptor in mice were examined in two severity grades of cecal ligation and puncture (CLP) leading to mild sepsis or septic shock.
Results
Twenty hours after induction of the high grade CLP, clopidogrel- and vehicle-treated mice displayed a similar 30% decrease in mean arterial blood pressure (MAP) characteristic of shock. Septic shock-induced thrombocytopenia was not modified by clopidogrel treatment. Plasma concentrations of inflammatory cytokines and myeloperoxidase (MPO) were similarly increased in clopidogrel- and vehicle-treated mice, indicating comparable increase in systemic inflammation. Thrombin-antithrombin (TAT) complexes and the extent of organ damage were also similar. In mild-grade CLP, clopidogrel- and vehicle-treated mice did not display a significant decrease in MAP, while thrombocytopenia and plasma concentrations of TNFα, IL6, IL10, MPO, TAT and organ damage reached similar levels in both groups, although lower than those reached in the high grade CLP. Similarly, mice with platelet-specific deletion of the P2Y
12
receptor were not protected from CLP-induced sepsis or septic shock.
Conclusion
The platelet P2Y
12
receptor does not contribute to the pathogenesis of sepsis or septic shock in mice, suggesting that P2Y
12
receptor antagonists may not be beneficial in patients with sepsis or septic shock.
Collapse
Affiliation(s)
- Yannick Rabouel
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS)-Grand Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), F-67000 Strasbourg, France
| | - Stéphanie Magnenat
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS)-Grand Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), F-67000 Strasbourg, France
| | - Xavier Delabranche
- Hôpitaux Universitaires de Strasbourg, Anesthésie, Réanimation et Médecine périopératoire, Nouvel Hôpital Civil, F-67000 Strasbourg, France
| | - Christian Gachet
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS)-Grand Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), F-67000 Strasbourg, France
| | - Beatrice Hechler
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS)-Grand Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), F-67000 Strasbourg, France
| |
Collapse
|
40
|
Wang L, Su W, Xue J, Gong X, Dai Y, Chen J, Xue L, He P, Liu Y, Tan N. Association of thrombocytopenia and infection in patients with ST-elevation myocardial infarction undergoing percutaneous coronary intervention. BMC Cardiovasc Disord 2021; 21:404. [PMID: 34418967 PMCID: PMC8379583 DOI: 10.1186/s12872-021-02210-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 08/16/2021] [Indexed: 11/20/2022] Open
Abstract
Background The impact of thrombocytopenia on infection in patients with ST-elevation myocardial infarction (STEMI) remains poorly understood. Aims To evaluate the association between thrombocytopenia and infection in patients with STEMI. Methods Patients diagnosed with STEMI were identified from January 2010 to June 2016. The primary endpoint was in-hospital infection, and major adverse clinical events (MACE) and all-cause death were considered as secondary endpoints. Results A total of 1401 STEMI patients were enrolled and divided into two groups according to the presence (n = 186) or absence (n = 1215) of thrombocytopenia. The prevalence of in-hospital infection was significantly higher in the thrombocytopenic group (30.6% (57/186) vs. 16.2% (197/1215), p < 0.001). Prevalence of in-hospital MACE (30.1% (56/186) vs. 16.4% (199/1215), p < 0.001) and all-cause death (8.1% (15/186) vs. 3.8% (46/1215), p = 0.008) revealed an increasing trend. Multivariate analysis indicated that thrombocytopenia was independently associated with increased in-hospital infection (OR, 2.09; 95%CI 1.32–3.27; p = 0.001) and MACE (1.92; 1.27–2.87; p = 0.002), but not all-cause death (1.87; 0.88–3.78; p = 0.091). After a median follow-up of 2.85 years, thrombocytopenia was not associated with all-cause death at multivariable analysis (adjusted hazard ratio, 1.19; 95%CI 0.80–1.77; p = 0.383). Conclusions Thrombocytopenia is significantly correlated with in-hospital infection and MACE, and might be used as a prognostic tool in patients with STEMI. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-021-02210-3.
Collapse
Affiliation(s)
- Litao Wang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, China.,Guangdong Provincial People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510100, China
| | - Weijiang Su
- Department of Cardiology, The People's Hospital of Dianbai District, Maoming, 525400, China
| | - Jinhua Xue
- Department of Physiology, School of Basic Medical Sciences, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, 341000, China
| | - Xiao Gong
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yining Dai
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, China
| | - Jiyan Chen
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, China
| | - Ling Xue
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, China
| | - Pengcheng He
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, China.,Guangdong Provincial People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510100, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yuanhui Liu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, China.
| | - Ning Tan
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, China. .,Guangdong Provincial People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510100, China. .,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
41
|
Na YR, Jung D, Song J, Park JW, Hong JJ, Seok SH. Pyruvate dehydrogenase kinase is a negative regulator of interleukin-10 production in macrophages. J Mol Cell Biol 2021; 12:543-555. [PMID: 31900478 PMCID: PMC7493030 DOI: 10.1093/jmcb/mjz113] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/11/2019] [Accepted: 11/09/2019] [Indexed: 02/07/2023] Open
Abstract
Interleukin-10 (IL-10) is the most potent anti-inflammatory cytokine in the body and plays an essential role in determining outcomes of many inflammatory diseases. Cellular metabolism is a critical determinant of immune cell function; however, it is currently unclear whether metabolic processes are specifically involved in IL-10 production. In this study, we aimed to find the central metabolic molecule regulating IL-10 production of macrophages, which are the main producers of IL-10. Transcriptomic analysis identified that metabolic changes were predominantly enriched in Kupffer cells at the early inflammatory phase of a mouse endotoxemia model. Among them, pyruvate dehydrogenase kinase (PDK)-dependent acute glycolysis was negatively involved in IL-10 production. Inhibition or knockdown of PDK selectively increased macrophage IL-10 expression. Mechanistically, PDK inhibition increased IL-10 production via profound phosphorylation of adenosine monophosphate (AMP)-activated protein kinase alpha 1 (AMPKα1) by restricting glucose uptake in lipopolysaccharide-stimulated macrophages. AMPKα1 consequently activated p38 mitogen-activated protein kinase, c-Jun N-terminal kinase, and cyclic AMP-responsive element-binding protein to regulate IL-10 production. Our study uncovers a previously unknown regulatory mechanism of IL-10 in activated macrophages involving an immunometabolic function of PDK.
Collapse
Affiliation(s)
- Yi Rang Na
- Macrophage Lab, Department of Microbiology and Immunology and Institute of Endemic Disease, Seoul National University College of Medicine, Chongno-gu, Seoul 03080, Republic of Korea
| | - Daun Jung
- Macrophage Lab, Department of Microbiology and Immunology and Institute of Endemic Disease, Seoul National University College of Medicine, Chongno-gu, Seoul 03080, Republic of Korea
| | - Juha Song
- Macrophage Lab, Department of Microbiology and Immunology and Institute of Endemic Disease, Seoul National University College of Medicine, Chongno-gu, Seoul 03080, Republic of Korea
| | - Jong-Wan Park
- Department of Biomedical Science, BK21-Plus Education Program, Seoul National University College of Medicine, Jongno-gu, Seoul 03080, Republic of Korea.,Department of Pharmacology, Seoul National University College of Medicine, Jongno-gu, Seoul 03080, Republic of Korea.,Cancer Research Institute and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Jongno-gu, Seoul 03080, Republic of Korea
| | - Jung Joo Hong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Seung Hyeok Seok
- Macrophage Lab, Department of Microbiology and Immunology and Institute of Endemic Disease, Seoul National University College of Medicine, Chongno-gu, Seoul 03080, Republic of Korea
| |
Collapse
|
42
|
Han YC, Xie HZ, Lu B, Xiang RL, Zhang HP, Li JY, Zhang SY. Lipopolysaccharide Alters the m6A Epitranscriptomic Tagging of RNAs in Cardiac Tissue. Front Mol Biosci 2021; 8:670160. [PMID: 34395520 PMCID: PMC8355517 DOI: 10.3389/fmolb.2021.670160] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 07/14/2021] [Indexed: 11/13/2022] Open
Abstract
N6-methyladenosine (m6A) modification plays important roles in the pathology of a variety of diseases. However, the roles of m6A modification in sepsis-induced myocardial dysfunction are not well defined. Rats were divided into control and lipopolysaccharide (LPS)-induced sepsis group. Global m6A levels of left ventricle tissue were measured by LC-MS/MS, and transcriptome-wide m6A modifications were profiled using epitranscriptomic microarrays (mRNAs and lncRNAs). Bioinformatics analysis was conducted to understand the functional implications of m6A modifications during sepsis. Methylated lncRNAs and mRNAs were measured by m6A single-base site qPCR. The global m6A levels in left ventricle tissue were significantly decreased in the LPS group. While 27 transcripts (23 mRNAs and four lncRNAs) were hypermethylated, 46 transcripts (39 mRNAs and 7 lncRNAs) were hypomethylated in the LPS group. The mRNA expression of writers and readers was significantly decreased in the LPS group. The m6A modification of Clec1b, Stk38l and Tnfrsf26 was associated with platelet activation and apoptotic pathways. Moreover, the decrease in m6A modification of lncRNA XR_346,771 may be related to cation import in cardiac tissue. Our data provide novel information regarding changes to m6A modifications in cardiac tissue during sepsis, and m6A modifications might be promising therapeutic targets.
Collapse
Affiliation(s)
- Ye-Chen Han
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong-Zhi Xie
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Lu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruo-Lan Xiang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Beijing, China
| | - Hai-Peng Zhang
- Peking University Fifth School of Clinical Medicine (Beijing Hospital), Beijing, China
| | - Jing-Yi Li
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shu-Yang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
43
|
Yang S, Huang X, Liao J, Li Q, Chen S, Liu C, Ling L, Zhou J. Platelet-leukocyte aggregates - a predictor for acute kidney injury after cardiac surgery. Ren Fail 2021; 43:1155-1162. [PMID: 34266358 PMCID: PMC8288121 DOI: 10.1080/0886022x.2021.1948864] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background Acute kidney injury (AKI) is one of the most common complications after cardiac surgery. However, effective biomarker used for early diagnosis of AKI has not been identified. Platelet-leukocyte aggregates (PLAs) participate in inflammation and coagulation, leading to vascular lesions and tissue destruction. We designed a prospective study to assess whether PLAs can serve as a good biomarker for early diagnosis of AKI after cardiac surgery. Methods Patients with rheumatic heart disease scheduled to undergo valve replacement surgery were enrolled. Blood samples were collected at five timepoints as follows: (a) At baseline. (b) At the end of extracorporeal circulation. (c) Arrival at intensive care unit (ICU). (d) Four-hours after the admission to ICU. (e) Twenty hours after the admission to ICU. After collection, the samples were immediately used for PLAs measurement by flow cytometry. Results A total of 244 patients were registered, and 15 of them were diagnosed with AKI according to the serum creatinine of KDIGO guidelines. The PLAs levels in AKI group were significantly increased 20 h after surgery (two-way repeated measure analysis of variance, p < 0.01) compared with that at baseline. Patients whose preoperative PLAs were higher than 6.8% showed increased risk of developing AKI (multivariate logistic regression; p = 0.01; adjusted odds ratio, 1.05; 95% confidence interval, 1.01–1.09). Conclusion PLAs is an independent risk factor for AKI after valve replacement among patients with rheumatic heart disease.
Collapse
Affiliation(s)
- Shenghan Yang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xunbei Huang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Juan Liao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Qin Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Si Chen
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Chaonan Liu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Liqin Ling
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
44
|
Bourne JH, Beristain-Covarrubias N, Zuidscherwoude M, Campos J, Di Y, Garlick E, Colicchia M, Terry LV, Thomas SG, Brill A, Bayry J, Watson SP, Rayes J. CLEC-2 Prevents Accumulation and Retention of Inflammatory Macrophages During Murine Peritonitis. Front Immunol 2021; 12:693974. [PMID: 34163489 PMCID: PMC8215360 DOI: 10.3389/fimmu.2021.693974] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/20/2021] [Indexed: 11/29/2022] Open
Abstract
Platelets play a key role in the development, progression and resolution of the inflammatory response during sterile inflammation and infection, although the mechanism is not well understood. Here we show that platelet CLEC-2 reduces tissue inflammation by regulating inflammatory macrophage activation and trafficking from the inflamed tissues. The immune regulatory function of CLEC-2 depends on the expression of its ligand, podoplanin, upregulated on inflammatory macrophages and is independent of platelet activation and secretion. Mechanistically, platelet CLEC-2 and also recombinant CLEC-2-Fc accelerates actin rearrangement and macrophage migration by increasing the expression of podoplanin and CD44, and their interaction with the ERM proteins. During ongoing inflammation, induced by lipopolysaccharide, treatment with rCLEC-2-Fc induces the rapid emigration of peritoneal inflammatory macrophages to mesenteric lymph nodes, thus reducing the accumulation of inflammatory macrophages in the inflamed peritoneum. This is associated with a significant decrease in pro-inflammatory cytokine, TNF-α and an increase in levels of immunosuppressive, IL-10 in the peritoneum. Increased podoplanin expression and actin remodelling favour macrophage migration towards CCL21, a soluble ligand for podoplanin and chemoattractant secreted by lymph node lymphatic endothelial cells. Macrophage efflux to draining lymph nodes induces T cell priming. In conclusion, we show that platelet CLEC-2 reduces the inflammatory phenotype of macrophages and their accumulation, leading to diminished tissue inflammation. These immunomodulatory functions of CLEC-2 are a novel strategy to reduce tissue inflammation and could be therapeutically exploited through rCLEC-2-Fc, to limit the progression to chronic inflammation.
Collapse
Affiliation(s)
- Joshua H. Bourne
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Nonantzin Beristain-Covarrubias
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Malou Zuidscherwoude
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| | - Joana Campos
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Ying Di
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Evelyn Garlick
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| | - Martina Colicchia
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Lauren V. Terry
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Steven G. Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| | - Alexander Brill
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe - Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, Paris, France
- Biological Sciences and Engineering, Indian Institute of Technology Palakkad, Kerala, India
| | - Steve P. Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| | - Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| |
Collapse
|
45
|
Rolfes V, Ribeiro LS, Hawwari I, Böttcher L, Rosero N, Maasewerd S, Santos MLS, Próchnicki T, Silva CMDS, Wanderley CWDS, Rothe M, Schmidt SV, Stunden HJ, Bertheloot D, Rivas MN, Fontes CJ, Carvalho LH, Cunha FQ, Latz E, Arditi M, Franklin BS. Platelets Fuel the Inflammasome Activation of Innate Immune Cells. Cell Rep 2021; 31:107615. [PMID: 32402278 PMCID: PMC7225754 DOI: 10.1016/j.celrep.2020.107615] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 03/12/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
The inflammasomes control the bioactivity of pro-inflammatory cytokines of the interleukin (IL)-1 family. The inflammasome assembled by NLRP3 has been predominantly studied in homogeneous cell populations in vitro, neglecting the influence of cellular interactions that occur in vivo. Here, we show that platelets boost the inflammasome capacity of human macrophages and neutrophils and are critical for IL-1 production by monocytes. Platelets license NLRP3 transcription, thereby enhancing ASC oligomerization, caspase-1 activity, and IL-1β secretion. Platelets influence IL-1β production in vivo, and blood platelet counts correlate with plasmatic IL-1β levels in malaria. Furthermore, we reveal an enriched platelet gene signature among the highest-expressed transcripts in IL-1β-driven autoinflammatory diseases. The platelet effect is independent of cell-to-cell contact, platelet-derived lipid mediators, purines, nucleic acids, and a host of platelet cytokines, and it involves the triggering of calcium-sensing receptors on macrophages. Hence, platelets provide an additional layer of regulation of inflammasomes and IL-1-driven inflammation. Platelets license NLRP3 for inflammasome activattion in innate immune cells Platelets are required for optimal monocyte inflammasome activation Platelets shape IL-1β in vivo, and platelet counts correlate with IL-1β in plasma A constitutive, heat-sensitive soluble platelet-factor boost IL-1β in macrophages
Collapse
Affiliation(s)
- Verena Rolfes
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Lucas Secchim Ribeiro
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany.
| | - Ibrahim Hawwari
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Lisa Böttcher
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Nathalia Rosero
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Salie Maasewerd
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Marina Lima Silva Santos
- Laboratório de Malária, Instituto René Rachou, Fundação Oswaldo Cruz, 30190-002 Belo Horizonte, MG, Brazil
| | - Tomasz Próchnicki
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Camila Meirelles de Souza Silva
- Center for Research in Inflammatory Diseases, School of Medicine of Ribeirão Preto, University of Sao Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Carlos Wagner de Souza Wanderley
- Center for Research in Inflammatory Diseases, School of Medicine of Ribeirão Preto, University of Sao Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Maximilian Rothe
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Susanne V Schmidt
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - H James Stunden
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Damien Bertheloot
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany
| | - Magali Noval Rivas
- Departments of Pediatrics, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Cor Jesus Fontes
- Departamento de Clínica Médica, Universidade Federal de Mato Grosso, 78060-900 Cuiabá, MT, Brazil
| | - Luzia Helena Carvalho
- Laboratório de Malária, Instituto René Rachou, Fundação Oswaldo Cruz, 30190-002 Belo Horizonte, MG, Brazil
| | - Fernando Queiroz Cunha
- Center for Research in Inflammatory Diseases, School of Medicine of Ribeirão Preto, University of Sao Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Eicke Latz
- Institute of Innate Immunity, Medical Faculty, University of Bonn, 53127 Bonn, NRW, Germany; Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01655, USA; German Center for Neurodegenerative Diseases, 53127 Bonn, NRW, Germany
| | - Moshe Arditi
- Departments of Pediatrics, Division of Infectious Diseases and Immunology, and Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA.
| | | |
Collapse
|
46
|
Heffron SP, Weinstock A, Scolaro B, Chen S, Sansbury BE, Marecki G, Rolling CC, El Bannoudi H, Barrett T, Canary JW, Spite M, Berger JS, Fisher EA. Platelet-conditioned media induces an anti-inflammatory macrophage phenotype through EP4. J Thromb Haemost 2021; 19:562-573. [PMID: 33171016 PMCID: PMC7902474 DOI: 10.1111/jth.15172] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 10/24/2020] [Accepted: 10/27/2020] [Indexed: 02/02/2023]
Abstract
BACKGROUND Platelets are increasingly recognized as immune cells. As such, they are commonly seen to induce and perpetuate inflammation; however, anti-inflammatory activities are increasingly attributed to them. Atherosclerosis is a chronic inflammatory condition. Similar to other inflammatory conditions, the resolution of atherosclerosis requires a shift in macrophages to an M2 phenotype, enhancing their efferocytosis and cholesterol efflux capabilities. OBJECTIVES To assess the effect of platelets on macrophage phenotype. METHODS In several in vitro models employing murine (RAW264.7 and bone marrow-derived macrophages) and human (THP-1 and monocyte-derived macrophages) cells, we exposed macrophages to media in which non-agonized human platelets were cultured for 60 minutes (platelet-conditioned media [PCM]) and assessed the impact on macrophage phenotype and function. RESULTS Across models, we demonstrated that PCM from healthy humans induced a pro-resolving phenotype in macrophages. This was independent of signal transducer and activator of transcription 6 (STAT6), the prototypical pathway for M2 macrophage polarization. Stimulation of the EP4 receptor on macrophages by prostaglandin E2 present in PCM, is at least partially responsible for altered gene expression and associated function of the macrophages-specifically reduced peroxynitrite production, increased efferocytosis and cholesterol efflux capacity, and increased production of pro-resolving lipid mediators (ie, 15R-LXA4 ). CONCLUSIONS Platelet-conditioned media induces an anti-inflammatory, pro-resolving phenotype in macrophages. Our findings suggest that therapies targeting hemostatic properties of platelets, while not influencing pro-resolving, immune-related activities, could be beneficial for the treatment of atherothrombotic disease.
Collapse
Affiliation(s)
- Sean P. Heffron
- NYU Langone Health, Leon H. Charney Division of Cardiology, New York, NY, USA
- NYU Langone Health, NYU Center for the Prevention of Cardiovascular Disease, New York, NY, USA
| | - Ada Weinstock
- NYU Langone Health, Leon H. Charney Division of Cardiology, New York, NY, USA
| | - Bianca Scolaro
- NYU Langone Health, Leon H. Charney Division of Cardiology, New York, NY, USA
| | - Shiyu Chen
- NYU Department of Chemistry, New York, NY, USA
| | - Brian E. Sansbury
- Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Greg Marecki
- NYU Langone Health, Leon H. Charney Division of Cardiology, New York, NY, USA
| | | | - Hanane El Bannoudi
- NYU Langone Health, Leon H. Charney Division of Cardiology, New York, NY, USA
| | - Tessa Barrett
- NYU Langone Health, Leon H. Charney Division of Cardiology, New York, NY, USA
| | | | - Matthew Spite
- Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Jeffrey S. Berger
- NYU Langone Health, Leon H. Charney Division of Cardiology, New York, NY, USA
- NYU Langone Health, NYU Center for the Prevention of Cardiovascular Disease, New York, NY, USA
- NYU Langone Health, Department of Surgery, New York University, New York, NY, USA
| | - Edward A. Fisher
- NYU Langone Health, Leon H. Charney Division of Cardiology, New York, NY, USA
- NYU Langone Health, NYU Center for the Prevention of Cardiovascular Disease, New York, NY, USA
| |
Collapse
|
47
|
Zhu Y, Chen X, Lu Y, Fan S, Yang Y, Chen Q, Huang Q, Xia L, Wei Y, Zheng J, Liu X. Diphenyleneiodonium enhances P2X7 dependent non-opsonized phagocytosis and suppresses inflammasome activation via blocking CX43-mediated ATP leakage. Pharmacol Res 2021; 166:105470. [PMID: 33529751 DOI: 10.1016/j.phrs.2021.105470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/20/2021] [Accepted: 01/25/2021] [Indexed: 12/20/2022]
Abstract
The beneficial effects of antioxidants against oxidative stress have been well described. However, the pharmacological impacts of antioxidants other than inhibiting the production of reactive oxygen species (ROS) remain less understood. This study demonstrated that diphenyleneiodonium (DPI), a canonical NADPH oxidase 2 (NOX2) inhibitor, effectively promoted non-opsonized bacterial phagocytosis. Indeed, DPI abrogated the elevation in the extracellular ATP level of Escherichia coli (E. coli) -infected murine peritoneal macrophages, thereby restoring the association of the purinergic receptor P2X7 with non-muscle myosin heavy chain 9 (MYH9) to upregulate the P2X7 -dependent phagocytosis of E. coli. DPI also suppressed inflammasome activation and reduced necroptosis in E. coli-infected macrophages by decreasing extracellular ATP levels. Mechanistically, DPI upregulated p38 MAPK phosphorylation to suppress the expression and activity of the hemichannel protein connexin 43 (CX43), leading to the inhibition of CX43-mediated ATP efflux in E. coli-infected macrophages. In a murine E. coli infection model, DPI effectively reduced ATP release, decreased bacterial load and inhibited inflammasome activation, thereby improving survival and ameliorating organ injuries in model mice. In summary, our study demonstrates a previously unknown function of DPI in conferring protection against bacterial infection and suggests a putative antimicrobial strategy of modulating CX43 -dependent ATP leakage.
Collapse
Affiliation(s)
- Yuanfeng Zhu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiaoli Chen
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yongling Lu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shijun Fan
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yongjun Yang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qian Chen
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qianying Huang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lin Xia
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yan Wei
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jiang Zheng
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xin Liu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
48
|
Sheyn D, Darvish R, Nayak L, Myer S, Claridge C, Bretschneider CE. Perioperative outcomes for benign hysterectomy among women with thrombocytopenia. Int J Gynaecol Obstet 2021; 154:233-240. [PMID: 33420719 DOI: 10.1002/ijgo.13582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/28/2020] [Accepted: 01/05/2021] [Indexed: 01/19/2023]
Abstract
OBJECTIVE To determine whether mild or moderate thrombocytopenia is associated with postoperative complications after benign hysterectomy. METHODS A retrospective study of data from women who underwent benign hysterectomy included in the American College of Surgeons National Surgical Quality Improvement Project Database. The data were stratified by normal platelet count, mild thrombocytopenia (100-149 × 103 platelets/µl), and moderate thrombocytopenia (50-99 × 103 platelets/µl). Multivariable logistic regression was used to determine the relationship between mild or moderate thrombocytopenia and the main outcome measures. RESULTS Moderate thrombocytopenia was associated with an increased risk of perioperative transfusion (adjusted odds ratio [aOR], 2.87; 95% confidence interval [CI], 1.96-4.21) and reoperation (aOR, 4.03; 95% CI, 1.94-17.33), but mild thrombocytopenia was not. There was an increased risk of infection among women with both mild (aOR, 1.38; 95% CI, 1.12-1.69) and moderate (aOR, 2.00; 95% CI,1.23-3.22) thrombocytopenia. There was no association between either mild or moderate thrombocytopenia and readmission, prolonged hospital stay, or longer surgical time. CONCLUSION Thrombocytopenia was found to be associated with increased infectious morbidity after hysterectomy, and moderate thrombocytopenia was associated with an increased risk of perioperative transfusion and reoperation.
Collapse
Affiliation(s)
- David Sheyn
- Division of Female Pelvic Medicine and Reconstructive Surgery, Department of Obstetrics and Gynecology, MetroHealth Medical Center, Cleveland, OH, USA.,Department of Obstetrics and Gynecology, MetroHealth Medical Center, Cleveland, OH, USA.,Section of Urogynecology and Reconstructive Pelvic Surgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Ryan Darvish
- Department of Obstetrics and Gynecology, MetroHealth Medical Center, Cleveland, OH, USA.,Section of Urogynecology and Reconstructive Pelvic Surgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Lalitha Nayak
- Section of Urogynecology and Reconstructive Pelvic Surgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,Division of Hematology & Oncology, Department of Internal Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Sara Myer
- Section of Urogynecology and Reconstructive Pelvic Surgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Caitlin Claridge
- Department of Obstetrics and Gynecology, MetroHealth Medical Center, Cleveland, OH, USA.,Section of Urogynecology and Reconstructive Pelvic Surgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - C Emi Bretschneider
- Division of Female Pelvic Medicine and Reconstructive Surgery, Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL, USA
| |
Collapse
|
49
|
Nabil-Adam A, Shreadah MA. Anti-inflammatory, Antioxidant, Lung and Liver Protective Activity of Galaxaura oblongata as Antagonistic Efficacy against LPS using Hematological Parameters and Immunohistochemistry as Biomarkers. Cardiovasc Hematol Agents Med Chem 2021; 20:148-165. [PMID: 33438570 DOI: 10.2174/1871525719666210112154800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 10/28/2020] [Accepted: 11/16/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND This study aimed to investigate the potential bioactivity and the ameliorative role of Galaxaura oblongata (G. oblongata) against LPS-induced toxicity by using hematological parameters. OBJECTIVE It is aimed also to examine its protective effect using the immunohistochemistry of liver and lungs as biomarkers in male BALB/C albino mice. MATERIALS AND METHODS The current study carried out using different in-vitro and in-vivo assays such as phytochemical, antioxidants, anti-inflammatory for in-vitro where the hematological and immunohistochemistry for lung and liver were investigated in vivo. RESULTS There are no previous studies were performed to investigate the in vivo and in vitro effects of the G. oblongata extracts as antioxidant and anti-inflammatory due to their rareness compared to other red algae. LPS treated mice revealed a significant decrease in total number of WBCs, RBCs, platelets, and HGB%, MPV, MCV and MCHC compared to the control group. On contrast, the HCT and MCHC were increased in the induction group which was treated with LPS compared to the control group. Furthermore, the immunohistochemistry results of the present study revealed the protective effect of G. oblongata compared to the induction group. G. oblongata can be used as protective marine natural products against the toxicity induced by LPS. CONCLUSION It exhibited a significant ameliorative role against the alterations in the hematological parameters and immunohistochemistry of liver and lungs, and helps to reduce as well as coordinate the acute inflammations caused by TNF.
Collapse
Affiliation(s)
- Asmaa Nabil-Adam
- Marine Biotechnology and Natural Products Lab (MBNP), National Institute of Oceanography & Fisheries (NIOF), Alexandria. Egypt
| | - Mohamed A Shreadah
- Marine Biotechnology and Natural Products Lab (MBNP), National Institute of Oceanography & Fisheries (NIOF), Alexandria. Egypt
| |
Collapse
|
50
|
Wang Y, Chen W, Zhang W, Lee-Sundlov MM, Casari C, Berndt MC, Lanza F, Bergmeier W, Hoffmeister KM, Zhang XF, Li R. Desialylation of O-glycans on glycoprotein Ibα drives receptor signaling and platelet clearance. Haematologica 2021; 106:220-229. [PMID: 31974202 PMCID: PMC7776245 DOI: 10.3324/haematol.2019.240440] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 01/22/2020] [Indexed: 11/16/2022] Open
Abstract
During infection neuraminidase desialylates platelets and induces their rapid clearance from circulation. The underlying molecular basis, particularly the role of platelet glycoprotein (GP)Ibα therein, is not clear. Utilizing genetically altered mice, we report that the extracellular domain of GPIbα, but neither von Willebrand factor nor ADAM17 (a disintegrin and metalloprotease 17), is required for platelet clearance induced by intravenous injection of neuraminidase. Lectin binding to platelet following neuraminidase injection over time revealed that the extent of desialylation of O-glycans correlates with the decrease of platelet count in mice. Injection of α2,3-neuraminidase reduces platelet counts in wild-type but not in transgenic mice expressing only a chimeric GPIbα that misses most of its extracellular domain. Neuraminidase treatment induces unfolding of the O-glycosylated mechanosensory domain in GPIbα as monitored by single-molecule force spectroscopy, increases the exposure of the ADAM17 shedding cleavage site in the mechanosensory domain on the platelet surface, and induces ligand-independent GPIb-IX signaling in human and murine platelets. These results suggest that desialylation of O-glycans of GPIbα induces unfolding of the mechanosensory domain, subsequent GPIb-IX signaling including amplified desialylation of N-glycans, and eventually rapid platelet clearance. This new molecular mechanism of GPIbα-facilitated clearance could potentially resolve many puzzling and seemingly contradicting observations associated with clearance of desialylated or hyposialylated platelet.
Collapse
Affiliation(s)
- Yingchun Wang
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Wenchun Chen
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Wei Zhang
- Department of Bioengineering, Lehigh University, Bethlehem, PA
| | | | - Caterina Casari
- McAllister Heart Institute, University of North Carolina, School of Medicine, Chapel Hill, NC
| | | | - Francois Lanza
- Université de Strasbourg, EFS-Alsace, Strasbourg, France
| | | | | | - X Frank Zhang
- Department of Bioengineering, Lehigh University, Bethlehem, PA
| | - Renhao Li
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|