1
|
Sun P, Cui M, Jing J, Kong F, Wang S, Tang L, Leng J, Chen K. Deciphering the molecular and cellular atlas of immune cells in septic patients with different bacterial infections. J Transl Med 2023; 21:777. [PMID: 37919720 PMCID: PMC10621118 DOI: 10.1186/s12967-023-04631-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/14/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Sepsis is a life-threatening organ dysfunction caused by abnormal immune responses to various, predominantly bacterial, infections. Different bacterial infections lead to substantial variation in disease manifestation and therapeutic strategies. However, the underlying cellular heterogeneity and mechanisms involved remain poorly understood. METHODS Multiple bulk transcriptome datasets from septic patients with 12 types of bacterial infections were integrated to identify signature genes for each infection. Signature genes were mapped onto an integrated large single-cell RNA (scRNA) dataset from septic patients, to identify subsets of cells associated with different sepsis types, and multiple omics datasets were combined to reveal the underlying molecular mechanisms. In addition, an scRNA dataset and spatial transcriptome data were used to identify signaling pathways in sepsis-related cells. Finally, molecular screening, optimization, and de novo design were conducted to identify potential targeted drugs and compounds. RESULTS We elucidated the cellular heterogeneity among septic patients with different bacterial infections. In Escherichia coli (E. coli) sepsis, 19 signature genes involved in epigenetic regulation and metabolism were identified, of which DRAM1 was demonstrated to promote autophagy and glycolysis in response to E. coli infection. DRAM1 upregulation was confirmed in an independent sepsis cohort. Further, we showed that DRAM1 could maintain survival of a pro-inflammatory monocyte subset, C10_ULK1, which induces systemic inflammation by interacting with other cell subsets via resistin and integrin signaling pathways in blood and kidney tissue, respectively. Finally, retapamulin was identified and optimized as a potential drug for treatment of E. coli sepsis targeting the signature gene, DRAM1, and inhibiting E. coli protein synthesis. Several other targeted drugs were also identified in other types of sepsis, including nystatin targeting C1QA in Neisseria sepsis and dalfopristin targeting CTSD in Streptococcus viridans sepsis. CONCLUSION Our study provides a comprehensive overview of the cellular heterogeneity and underlying mechanisms in septic patients with various bacterial infections, providing insights to inform development of stratified targeted therapies for sepsis.
Collapse
Affiliation(s)
- Ping Sun
- Translational Medical Center for Stem Cell Therapy, Institute for Regenerative Medicine, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, 200127, China
- Department of Emergency, Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Mintian Cui
- Translational Medical Center for Stem Cell Therapy, Institute for Regenerative Medicine, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, 200127, China
| | - Jiongjie Jing
- Translational Medical Center for Stem Cell Therapy, Institute for Regenerative Medicine, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, 200127, China
| | - Fanyu Kong
- Department of Internal Emergency Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Shixi Wang
- Translational Medical Center for Stem Cell Therapy, Institute for Regenerative Medicine, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, 200127, China
| | - Lunxian Tang
- Department of Internal Emergency Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Junling Leng
- Department of Emergency, Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Kun Chen
- Translational Medical Center for Stem Cell Therapy, Institute for Regenerative Medicine, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, 200127, China.
- Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
2
|
Trivigno SMG, Guidetti GF, Barbieri SS, Zarà M. Blood Platelets in Infection: The Multiple Roles of the Platelet Signalling Machinery. Int J Mol Sci 2023; 24:ijms24087462. [PMID: 37108623 PMCID: PMC10138547 DOI: 10.3390/ijms24087462] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Platelets are classically recognized for their important role in hemostasis and thrombosis but they are also involved in many other physiological and pathophysiological processes, including infection. Platelets are among the first cells recruited to sites of inflammation and infection and they exert their antimicrobial response actively cooperating with the immune system. This review aims to summarize the current knowledge on platelet receptor interaction with different types of pathogens and the consequent modulations of innate and adaptive immune responses.
Collapse
Affiliation(s)
- Silvia M G Trivigno
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
- University School for Advanced Studies, IUSS, 27100 Pavia, Italy
| | | | - Silvia Stella Barbieri
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy
| | - Marta Zarà
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy
| |
Collapse
|
3
|
Tuhongjiang A, Wang F, Zhang C, Pang S, Qu Y, Feng B, Amuti G. Construction of an RNA modification-related gene predictive model associated with prognosis and immunity in gastric cancer. BMC Bioinformatics 2023; 24:147. [PMID: 37061682 PMCID: PMC10105968 DOI: 10.1186/s12859-023-05283-3] [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: 01/25/2023] [Accepted: 04/12/2023] [Indexed: 04/17/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the most common causes of cancer-related fatalities worldwide, and its progression is associated with RNA modifications. Here, using RNA modification-related genes (RNAMRGs), we aimed to construct a prognostic model for patients with GC. METHODS Based on RNAMRGs, RNA modification scores (RNAMSs) were obtained for GC samples from The Cancer Genome Atlas and were divided into high- and low-RNAMS groups. Differential analysis and weighted correlation network analysis were performed for the differential expressed genes (DEGs) to obtain the key genes. Next, univariate Cox regression, least absolute shrinkage and selection operator, and multivariate Cox regression analyses were performed to obtain the model. According to the model risk score, samples were divided into high- and low-risk groups. Enrichment analysis and immunoassays were performed for the DEGs in these groups. Four external datasets from Gene Expression Omnibus data base were used to test the accuracy of the predictive model. RESULTS We identified SELP and CST2 as key DEGs, which were used to generate the predictive model. The high-risk group had a worse prognosis compared to the low-risk group (p < 0.05). Enrichment analysis and immunoassays revealed that 144 DEGs related to immune cell infiltration were associated with the Wnt signaling pathway and included hub genes such as ELN. Overall mutation levels, tumor mutation burden, and microsatellite instability were lower, but tumor immune dysfunction and exclusion scores were greater (p < 0.05) in the high-risk group than in the low-risk group. The validation results showed that the prediction model score can accurately predict the prognosis of GC patients. Finally, a nomogram was constructed using the risk score combined with the clinicopathological characteristics of patients with GC. CONCLUSION This risk score from the prediction model related to the tumor microenvironment and immunotherapy could accurately predict the overall survival of GC patients.
Collapse
Affiliation(s)
- Airexiati Tuhongjiang
- Department of Day Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, China
| | - Feng Wang
- Department of Day Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, China.
| | - Chengrong Zhang
- Department of Day Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, China
| | - Sisi Pang
- Department of Day Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, China
| | - Yujiang Qu
- Department of Day Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, China
| | - Bo Feng
- Department of Day Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, China
| | - Gulimire Amuti
- Department of Day Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, China
| |
Collapse
|
4
|
Cleary SJ, Conrad C. Investigating and imaging platelets in inflammation. Int J Biochem Cell Biol 2023; 157:106373. [PMID: 36716816 DOI: 10.1016/j.biocel.2023.106373] [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: 10/21/2022] [Revised: 01/12/2023] [Accepted: 01/23/2023] [Indexed: 01/29/2023]
Abstract
Blood platelets are best known for their roles in hemostasis and thrombosis, but platelets also make important contributions to inflammation, immunity, and inflammatory resolution. Experiments involving depletion, genetic modification, and live imaging of platelets in animal models have increased our mechanistic understanding of platelet contributions to inflammation. In this minireview, we provide a critical overview of experimental techniques for manipulating and imaging platelets in inflammation models. We then highlight studies using innovative approaches to elucidate molecular mechanisms through which platelet subsets, platelet Fc gamma receptors, and pro-resolution platelet functions influence inflammatory responses. We also propose future technologies and research directions which might move us closer to harnessing of platelet functions for improved therapeutic modulation of inflammatory diseases.
Collapse
Affiliation(s)
- Simon J Cleary
- Department of Medicine, UCSF, Health Sciences East 1355A, 513 Parnassus Ave., San Francisco, CA 94143, USA.
| | - Catharina Conrad
- Department of Medicine, UCSF, Health Sciences East 1355A, 513 Parnassus Ave., San Francisco, CA 94143, USA
| |
Collapse
|
5
|
Gautam I, Huss CW, Storad ZA, Krebs M, Bassiouni O, Ramesh R, Wuescher LM, Worth RG. Activated Platelets Mediate Monocyte Killing of Klebsiella pneumoniae. Infect Immun 2023; 91:e0055622. [PMID: 36853027 PMCID: PMC10016073 DOI: 10.1128/iai.00556-22] [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: 12/09/2022] [Accepted: 02/02/2023] [Indexed: 03/01/2023] Open
Abstract
Platelets are known for essential activities in hemostasis and for their important contribution to protection against infectious pathogens. Klebsiella pneumoniae is an opportunistic pathogen widely known to cause nosocomial infections. Recently, hypervirulent strains of K. pneumoniae have been emerging, which can cause severe infections in immunocompetent individuals. Combined with the increase in antibiotic resistance, it is important to understand how K. pneumoniae affects components of the immune system. We studied the interactions of human platelets with several K. pneumoniae strains (the wild type encapsulated strain, and a nonencapsulated mutant). Thrombin-stimulated whole human and mouse blood significantly inhibited bacterial growth compared to unstimulated whole blood. Furthermore, we investigated the effect of K. pneumoniae on platelet activation. Both strains induced significant increase in activation of both unstimulated and thrombin-stimulated human platelets. Additionally, only the nonencapsulated mutant increased aggregation of platelets in response to ADP. K. pneumoniae killing assays were then performed with washed platelets in the presence or absence of thrombin. Surprisingly, washed platelets failed to exhibit any effects on the growth of K. pneumoniae. We further explored the impact of platelets on monocyte-mediated killing of K. pneumoniae. Importantly, we found that activated platelets significantly enhanced monocyte-mediated killing of K. pneumoniae. This effect was likely due to the formation of platelet-monocyte aggregates in blood upon thrombin stimulation. Overall, this study highlights the role of platelets in mediating a protective response against K. pneumoniae and reinforces the importance of platelets in modulating leukocyte behavior.
Collapse
Affiliation(s)
- Iluja Gautam
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Chadwick W. Huss
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Zachary A. Storad
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Michelle Krebs
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Omar Bassiouni
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Rochan Ramesh
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Leah M. Wuescher
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| | - Randall G. Worth
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, Ohio, USA
| |
Collapse
|
6
|
Liao YE, Liu J, Arnold K. Heparan sulfates and heparan sulfate binding proteins in sepsis. Front Mol Biosci 2023; 10:1146685. [PMID: 36865384 PMCID: PMC9971734 DOI: 10.3389/fmolb.2023.1146685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Heparan sulfates (HSs) are the main components in the glycocalyx which covers endothelial cells and modulates vascular homeostasis through interactions with multiple Heparan sulfate binding proteins (HSBPs). During sepsis, heparanase increases and induces HS shedding. The process causes glycocalyx degradation, exacerbating inflammation and coagulation in sepsis. The circulating heparan sulfate fragments may serve as a host defense system by neutralizing dysregulated Heparan sulfate binding proteins or pro-inflammatory molecules in certain circumstances. Understanding heparan sulfates and heparan sulfate binding proteins in health and sepsis is critical to decipher the dysregulated host response in sepsis and advance drug development. In this review, we will overview the current understanding of HS in glycocalyx under septic condition and the dysfunctional heparan sulfate binding proteins as potential drug targets, particularly, high mobility group box 1 (HMGB1) and histones. Moreover, several drug candidates based on heparan sulfates or related to heparan sulfates, such as heparanase inhibitors or heparin-binding protein (HBP), will be discussed regarding their recent advances. By applying chemical or chemoenzymatic approaches, the structure-function relationship between heparan sulfates and heparan sulfate binding proteins is recently revealed with structurally defined heparan sulfates. Such homogenous heparan sulfates may further facilitate the investigation of the role of heparan sulfates in sepsis and the development of carbohydrate-based therapy.
Collapse
Affiliation(s)
- Yi-En Liao
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, United States
| | - Jian Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, United States
| | | |
Collapse
|
7
|
Schrottmaier WC, Kral-Pointner JB, Salzmann M, Mussbacher M, Schmuckenschlager A, Pirabe A, Brunnthaler L, Kuttke M, Maier B, Heber S, Datler H, Ekici Y, Niederreiter B, Heber U, Blomgren B, Gorki AD, Söderberg-Nauclér C, Payrastre B, Gratacap MP, Knapp S, Schabbauer G, Assinger A. Platelet p110β mediates platelet-leukocyte interaction and curtails bacterial dissemination in pneumococcal pneumonia. Cell Rep 2022; 41:111614. [PMID: 36351402 DOI: 10.1016/j.celrep.2022.111614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/15/2022] [Accepted: 10/14/2022] [Indexed: 11/09/2022] Open
Abstract
Phosphatidylinositol 3-kinase catalytic subunit p110β is involved in tumorigenesis and hemostasis. However, it remains unclear if p110β also regulates platelet-mediated immune responses, which could have important consequences for immune modulation during anti-cancer treatment with p110β inhibitors. Thus, we investigate how platelet p110β affects inflammation and infection. Using a mouse model of Streptococcus pneumoniae-induced pneumonia, we find that both platelet-specific p110β deficiency and pharmacologic inhibition of p110β with TGX-221 exacerbate disease pathogenesis by preventing platelet-monocyte and neutrophil interactions, diminishing their infiltration and enhancing bacterial dissemination. Platelet p110β mediates neutrophil phagocytosis of S. pneumoniae in vitro and curtails bacteremia in vivo. Genetic deficiency or inhibition of platelet p110β also impairs macrophage recruitment in an independent model of sterile peritonitis. Our results demonstrate that platelet p110β dysfunction exacerbates pulmonary infection by impeding leukocyte functions. Thereby, our findings provide important insights into the immunomodulatory potential of PI3K inhibitors in bacterial infection.
Collapse
Affiliation(s)
- Waltraud Cornelia Schrottmaier
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Julia Barbara Kral-Pointner
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Manuel Salzmann
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Marion Mussbacher
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria; Department of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, University of Graz, 8010 Graz, Austria
| | - Anna Schmuckenschlager
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Anita Pirabe
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Laura Brunnthaler
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Mario Kuttke
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Barbara Maier
- Department of Medicine I, Research Division of Infection Biology, Medical University Vienna, 1090 Vienna, Austria
| | - Stefan Heber
- Institute of Physiology, Centre for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Hannes Datler
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Yasemin Ekici
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Birgit Niederreiter
- Division of Rheumatology, Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria
| | - Ulrike Heber
- Department of Pathology and Comprehensive Cancer Centre, Medical University of Vienna, 1090 Vienna, Austria
| | - Bo Blomgren
- Department of Clinical Sciences, Danderyd Hospital, Department of Oncology-Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Anna-Dorothea Gorki
- Department of Medicine I, Research Division of Infection Biology, Medical University Vienna, 1090 Vienna, Austria
| | - Cecilia Söderberg-Nauclér
- Department of Medicine, Solna, Centre for Molecular Medicine, Microbial Pathogenesis Unit, Karolinska University Hospital, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Bernard Payrastre
- INSERM UMR1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Toulouse III Paul Sabatier, 31024 Toulouse, France
| | - Marie-Pierre Gratacap
- INSERM UMR1297, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Toulouse III Paul Sabatier, 31024 Toulouse, France
| | - Sylvia Knapp
- Department of Medicine I, Research Division of Infection Biology, Medical University Vienna, 1090 Vienna, Austria
| | - Gernot Schabbauer
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria.
| | - Alice Assinger
- Department of Vascular Biology and Thrombosis Research, Centre for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria.
| |
Collapse
|
8
|
Ludwig N, Hilger A, Zarbock A, Rossaint J. Platelets at the Crossroads of Pro-Inflammatory and Resolution Pathways during Inflammation. Cells 2022; 11:cells11121957. [PMID: 35741086 PMCID: PMC9221767 DOI: 10.3390/cells11121957] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 01/27/2023] Open
Abstract
Platelets are among the most abundant cells in the mammalian circulation. Classical platelet functions in hemostasis and wound healing have been intensively explored and are generally accepted. During the past decades, the research focus broadened towards their participation in immune-modulatory events, including pro-inflammatory and, more recently, inflammatory resolution processes. Platelets are equipped with a variety of abilities enabling active participation in immunological processes. Toll-like receptors mediate the recognition of pathogens, while the release of granule contents and microvesicles promotes direct pathogen defense and an interaction with leukocytes. Platelets communicate and physically interact with neutrophils, monocytes and a subset of lymphocytes via soluble mediators and surface adhesion receptors. This interaction promotes leukocyte recruitment, migration and extravasation, as well as the initiation of effector functions, such as the release of extracellular traps by neutrophils. Platelet-derived prostaglandin E2, C-type lectin-like receptor 2 and transforming growth factor β modulate inflammatory resolution processes by promoting the synthesis of pro-resolving mediators while reducing pro-inflammatory ones. Furthermore, platelets promote the differentiation of CD4+ T cells in T helper and regulatory T cells, which affects macrophage polarization. These abilities make platelets key players in inflammatory diseases such as pneumonia and the acute respiratory distress syndrome, including the pandemic coronavirus disease 2019. This review focuses on recent findings in platelet-mediated immunity during acute inflammation.
Collapse
|
9
|
The Underestimated Role of Platelets in Severe Infection a Narrative Review. Cells 2022; 11:cells11030424. [PMID: 35159235 PMCID: PMC8834344 DOI: 10.3390/cells11030424] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/16/2022] [Accepted: 01/23/2022] [Indexed: 12/13/2022] Open
Abstract
Beyond their role in hemostasis, platelets have emerged as key contributors in the immune response; accordingly, the occurrence of thrombocytopenia during sepsis/septic shock is a well-known risk factor of mortality and a marker of disease severity. Recently, some studies elucidated that the response of platelets to infections goes beyond a simple fall in platelets count; indeed, sepsis-induced thrombocytopenia can be associated with—or even anticipated by—several changes, including an altered morphological pattern, receptor expression and aggregation. Of note, alterations in platelet function and morphology can occur even with a normal platelet count and can modify, depending on the nature of the pathogen, the pattern of host response and the severity of the infection. The purpose of this review is to give an overview on the pathophysiological interaction between platelets and pathogens, as well as the clinical consequences of platelet dysregulation. Furthermore, we try to clarify how understanding the nature of platelet dysregulation may help to optimize the therapeutic approach.
Collapse
|
10
|
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: 14] [Impact Index Per Article: 3.5] [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
|
11
|
Asgari F, Supino D, Parente R, Polentarutti N, Stravalaci M, Porte R, Pasqualini F, Barbagallo M, Perucchini C, Recordati C, Magrini E, Mariancini A, Riva F, Giordano A, Davoudian S, Roger T, Veer CV, Jaillon S, Mantovani A, Doni A, Garlanda C. The Long Pentraxin PTX3 Controls Klebsiella Pneumoniae Severe Infection. Front Immunol 2021; 12:666198. [PMID: 34093560 PMCID: PMC8173212 DOI: 10.3389/fimmu.2021.666198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/04/2021] [Indexed: 12/29/2022] Open
Abstract
Klebsiella pneumoniae is a common pathogen in human sepsis. The emergence of multidrug-resistant K. pneumoniae strains represents a major clinical challenge in nosocomial and community acquired infections. The long pentraxin PTX3, a key component of humoral innate immunity, is involved in resistance to selected pathogens by promoting opsonophagocytosis. We investigated the relevance of PTX3 in innate immunity against K. pneumoniae infections using Ptx3-/- mice and mouse models of severe K. pneumoniae infections. Local and systemic PTX3 expression was induced following K. pneumoniae pulmonary infection, in association with the up-regulation of TNF-α and IL-1β. PTX3 deficiency in mice was associated with higher bacterial burden and mortality, release of pro-inflammatory cytokines as well as IL-10 in the lung and systemically. The analysis of the mechanisms responsible of PTX3-dependent control of K. pneumoniae infection revealed that PTX3 did not interact with K. pneumoniae, or promote opsonophagocytosis. The comparison of susceptibility of wild-type, Ptx3-/-, C3-/- and Ptx3-/-/C3-/- mice to the infection showed that PTX3 acted in a complement-independent manner. Lung histopathological analysis showed more severe lesions in Ptx3-/- mice with fibrinosuppurative, necrotizing and haemorrhagic bronchopneumonia, associated with increased fibrin deposition in the lung and circulating fibrinogen consumption. These findings indicate that PTX3 contributes to the control of K. pneumoniae infection by modulating inflammatory responses and tissue damage. Thus, this study emphasizes the relevance of the role of PTX3 as regulator of inflammation and orchestrator of tissue repair in innate responses to infections.
Collapse
Affiliation(s)
- Fatemeh Asgari
- Department of Inflammation and Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Domenico Supino
- Department of Inflammation and Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Raffaella Parente
- Department of Inflammation and Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Nadia Polentarutti
- Department of Inflammation and Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Matteo Stravalaci
- Department of Inflammation and Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Remi Porte
- Department of Inflammation and Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Fabio Pasqualini
- Department of Inflammation and Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Marialuisa Barbagallo
- Department of Inflammation and Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Chiara Perucchini
- Department of Inflammation and Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Camilla Recordati
- Department of Veterinary Medicine, University of Milano, Lodi, Italy
| | - Elena Magrini
- Department of Inflammation and Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Andrea Mariancini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Federica Riva
- Department of Veterinary Medicine, University of Milano, Lodi, Italy
| | - Alessia Giordano
- Department of Veterinary Medicine, University of Milano, Lodi, Italy
| | - Sadaf Davoudian
- Department of Inflammation and Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Cornelis Van't Veer
- Center of Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, Netherlands
| | - Sebastien Jaillon
- Department of Inflammation and Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Alberto Mantovani
- Department of Inflammation and Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy.,Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Andrea Doni
- Unit of Advanced Optical Microscopy, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Cecilia Garlanda
- Department of Inflammation and Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| |
Collapse
|
12
|
Karsli E, Sabirli R, Altintas E, Canacik O, Sabirli GT, Kaymaz B, Kurt Ö, Koseler A. Soluble P-selectin as a potential diagnostic and prognostic biomarker for COVID-19 disease: A case-control study. Life Sci 2021; 277:119634. [PMID: 34015287 PMCID: PMC8130002 DOI: 10.1016/j.lfs.2021.119634] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/02/2021] [Accepted: 05/08/2021] [Indexed: 12/11/2022]
Abstract
Introduction To our knowledge, the diagnostic value of the sP-Selectin level in the diagnosis of COVID-19 disease has not yet been investigated. In this study, we aimed to assess this by evaluating the relationship between sP-Selectin level and the clinical severity of COVID-19 infections. Methods A total of 80 patients (50 with mild to moderate and 30 with severe COVID-19 pneumonia), and 60 non-symptomatic healthy volunteers participated in the study. Following serum isolation, sP-Selectin levels were assessed by Enzyme-Linked Immunosorbent Assay (ELISA) method. Results The serum sP-Selectin level was 1.7 ng/ml in the control group (1–3.78); 6.24 ng/ml (5.14–7.23) in mild-to-moderate pneumonia group; and 6.72 ng/ml (5.36–8.03) in the severe pneumonia group. Serum sP-Selectin levels in both mild-to-moderate pneumonia and severe pneumonia groups were found to be higher than the control group, with statistical significance (p = 0.0001 and p = 0.0001, respectively). Receiver operating characteristic analysis (ROC) showed greater area under the curve (AUC) for the serum sP-Selectin levels of the COVID-19 patients (AUC = 0.913, 95% CI = 0.857–0.969; p = 0.0001). The serum sP-Selectin level was found to be 97.5% sensitive and 80% specific at 4.125 ng/ml level for diagnosis (p = 0.0001). The serum sP-Selectin level was found to be 76.9% sensitive and 51.9% specific at the level of 6.12 ng/ml (p = 0.005) to predict the need for intensive care treatment. Conclusion This study showed that sP-Selectin can be used as a valuable biomarker in both diagnosing and predicting the need for intensive care treatment of COVID-19 infection.
Collapse
Affiliation(s)
- Emre Karsli
- Department of Emergency Medicine, Kafkas University Faculty of Medicine, Kars, Turkey
| | - Ramazan Sabirli
- Department of Emergency Medicine, Kafkas University Faculty of Medicine, Kars, Turkey.
| | - Emel Altintas
- Department of Emergency Medicine, Ankara Training and Research Hospital, Ankara, Turkey
| | - Omer Canacik
- Department of Emergency Medicine, Kafkas University Faculty of Medicine, Kars, Turkey
| | | | - Buse Kaymaz
- Department of Medical Microbiology, Acibadem Mehmet Ali Aydinlar University School of Medicine, Istanbul, Turkey
| | - Özgür Kurt
- Department of Medical Microbiology, Acibadem Mehmet Ali Aydinlar University School of Medicine, Istanbul, Turkey
| | - Aylin Koseler
- Department of Biophysics, Pamukkale University Faculty of Medicine, Denizli, Turkey
| |
Collapse
|
13
|
Tanır Basaranoglu S, Ozsurekci Y, Aykac K, Iyigun I, Satirer O, Akin MS, Ceyhan M. Adhesion molecules as diagnostic and severity biomarkers in pediatric community-acquired pneumonia. CLINICAL RESPIRATORY JOURNAL 2021; 15:522-529. [PMID: 33484111 DOI: 10.1111/crj.13334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 09/09/2020] [Accepted: 01/19/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Discrimination of the cases with severe and mild pneumonia is crucial due to the requirement of hospitalization, additional management, and treatment protocols. We aimed to analyze the role of IL6 (Interleukin), IL8, IL10, VCAM-1 (soluble Vascular Cell Adhesion Molecule), and sSELE (soluble E-selectin) in the diagnosis and prognostic evaluation of community-acquired pneumonia (CAP). METHODS Pediatric patients with severe pneumonia (SP) were hospitalized and patients with mild disease (MP) were treated in the community. IL6, IL8, IL10, VCAM-1, and sSELE levels of the patients were investigated and compared with the age- and gender-matched healthy subjects. RESULTS A total of 113 patients fulfilling the criteria for a diagnosis of CAP were enrolled in the study, 62 (54.8%) of which had SP and 51 (45%) had MP. MP and SP groups were significantly different in terms of IL8, IL10, and sSELE levels. Patients with SP and MP had significantly different WBC, ESR, and CRP values, as well. CONCLUSIONS Besides classical acute phase parameters, inflammatory response parameters such as IL6 and VCAM-1 levels may be helpful in diagnosis of pneumonia. In terms of determination of disease severity in pediatric CAP, systemic inflammatory markers like IL8 and IL10 and adhesion molecules like sSELE seem useful in clinical settings.
Collapse
Affiliation(s)
| | - Yasemin Ozsurekci
- Pediatric Infectious Disease Department, Ihsan Dogramacı Children Hospital, Hacettepe University, Ankara, Turkey
| | - Kubra Aykac
- Ankara Training and Research Hospital, Ankara, Turkey
| | - Irem Iyigun
- Department of Pediatrics, Ihsan Dogramacı Children Hospital, Hacettepe University, Ankara, Turkey
| | - Ozlem Satirer
- Department of Pediatrics, Ihsan Dogramacı Children Hospital, Hacettepe University, Ankara, Turkey
| | - Mustafa Senol Akin
- Department of Pediatrics, Ihsan Dogramacı Children Hospital, Hacettepe University, Ankara, Turkey
| | - Mehmet Ceyhan
- Pediatric Infectious Disease Department, Ihsan Dogramacı Children Hospital, Hacettepe University, Ankara, Turkey
| |
Collapse
|
14
|
Rayes J, Bourne JH, Brill A, Watson SP. The dual role of platelet-innate immune cell interactions in thrombo-inflammation. Res Pract Thromb Haemost 2020; 4:23-35. [PMID: 31989082 PMCID: PMC6971330 DOI: 10.1002/rth2.12266] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/04/2019] [Accepted: 09/09/2019] [Indexed: 12/11/2022] Open
Abstract
Beyond their role in hemostasis and thrombosis, platelets are increasingly recognized as key regulators of the inflammatory response under sterile and infectious conditions. Both platelet receptors and secretion are critical for these functions and contribute to their interaction with the endothelium and innate immune system. Platelet-leukocyte interactions are increased in thrombo-inflammatory diseases and are sensitive biomarkers for platelet activation and targets for the development of new therapies. The crosstalk between platelets and innate immune cells promotes thrombosis, inflammation, and tissue damage. However, recent studies have shown that these interactions also regulate the resolution of inflammation, tissue repair, and wound healing. Many of the platelet and leukocyte receptors involved in these bidirectional interactions are not selective for a subset of immune cells. However, specific heterotypic interactions occur in different vascular beds and inflammatory conditions, raising the possibility of disease- and organ-specific pathways of intervention. In this review, we highlight and discuss prominent and emerging interrelationships between platelets and innate immune cells and their dual role in the regulation of the inflammatory response in sterile and infectious thrombo-inflammatory diseases. A better understanding of the functional relevance of these interactions in different vascular beds may provide opportunities for successful therapeutic interventions to regulate the development, progression, and chronicity of various pathological processes.
Collapse
Affiliation(s)
- Julie Rayes
- Institute of Cardiovascular SciencesCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)Universities of Birmingham and NottinghamThe MidlandsUK
| | - Joshua H. Bourne
- Institute of Cardiovascular SciencesCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Alexander Brill
- Institute of Cardiovascular SciencesCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)Universities of Birmingham and NottinghamThe MidlandsUK
- Department of PathophysiologySechenov First Moscow State Medical University (Sechenov University)MoscowRussia
| | - Steve P. Watson
- Institute of Cardiovascular SciencesCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Centre of Membrane Proteins and Receptors (COMPARE)Universities of Birmingham and NottinghamThe MidlandsUK
| |
Collapse
|
15
|
Li K, Yang C, Jiang Z, Liu S, Liu J, Fan C, Li T, Dong X. Quantitative investigation of factors relevant to the T cell spot test for tuberculosis infection in active tuberculosis. BMC Infect Dis 2019; 19:673. [PMID: 31357953 PMCID: PMC6664742 DOI: 10.1186/s12879-019-4310-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/23/2019] [Indexed: 02/03/2023] Open
Abstract
Background Previous qualitative studies suggested that the false negative rate of the T cell spot test for tuberculosis infection (T-SPOT.TB) is associated with many risk factors in tuberculosis patients. However, more precise quantitative studies are lacking. The purpose of this study was to investigate the factors affecting quantified spot-forming cells (SFCs) to early secreted antigenic target 6 kDa (ESAT-6) or culture filtrate protein 10 kDa (CFP-10) in patients with active tuberculosis. Methods We retrospectively analyzed the data of 360 patients who met the inclusion criteria. Using the SFCs to ESAT-6 or CFP-10 levels as dependent variables, variables with statistical significance in the univariate analysis were subjected to optimal scaling regression analysis. The combination of ESAT-6 and CFP-10 (i.e., T-SPOT.TB) was analyzed by the exact logistic regression model. Results The results showed that the SFCs to ESAT-6 regression model had statistical significance (P < 0.001) and that previous treatment and CD4+ and platelet counts were its independent risk factors (all P < 0.05). Their importance levels were 0.095, 0.596 and 0.100, respectively, with a total of 0.791. The SFCs to CFP-10 regression model also had statistical significance (P < 0.001); platelet distribution width and alpha-2 globulin were its independent risk factors (all P < 0.05). Their importance levels were 0.287 and 0.247, respectively, with a total of 0.534. The quantification graph showed that quantified SFCs to ESAT-6 or CFP-10 grading had a linear correlation with risk factors. Albumin-globulin ratio, CD4+ and CD8+ were independent risk factors for false negative T-SPOT.TB (all P < 0.05). Conclusions In T-SPOT.TB-assisted diagnosis of patients with active tuberculosis, previous treatment, decreased CD4+ and platelet count might lead to the decreased SFCs to ESAT-6, decreased alpha-2 globulin and high platelet distribution width might lead to the decreased SFCs to CFP-10, decreased albumin-globulin ratio, CD4+ and CD8+ might lead to an increase in the false negative rate of the T-SPOT.TB. Electronic supplementary material The online version of this article (10.1186/s12879-019-4310-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kui Li
- Department of Infectious Diseases, Ankang Central Hospital, 85 South Jinzhou Road, Ankang, 725000, Shaanxi Province, China.,Department of Infectious Diseases, Ankang Central Hospital, Hubei University of Medicine, 30 South Renmin Road, Shiyan, 442000, Hubei Province, China
| | - Caiyong Yang
- Department of Infectious Diseases, Ankang Central Hospital, 85 South Jinzhou Road, Ankang, 725000, Shaanxi Province, China. .,Department of Infectious Diseases, Ankang Central Hospital, Hubei University of Medicine, 30 South Renmin Road, Shiyan, 442000, Hubei Province, China.
| | - Zicheng Jiang
- Department of Infectious Diseases, Ankang Central Hospital, 85 South Jinzhou Road, Ankang, 725000, Shaanxi Province, China.,Department of Infectious Diseases, Ankang Central Hospital, Hubei University of Medicine, 30 South Renmin Road, Shiyan, 442000, Hubei Province, China
| | - Shengxi Liu
- Department of Infectious Diseases, Ankang Central Hospital, 85 South Jinzhou Road, Ankang, 725000, Shaanxi Province, China.,Department of Infectious Diseases, Ankang Central Hospital, Hubei University of Medicine, 30 South Renmin Road, Shiyan, 442000, Hubei Province, China
| | - Jun Liu
- Laboratory of Molecular Pathology and Tuberculosis Diseases, Ankang Central Hospital, 85 South Jinzhou Road, Ankang, 725000, Shaanxi Province, China
| | - Chuanqi Fan
- Department of Infectious Diseases, Ankang Central Hospital, 85 South Jinzhou Road, Ankang, 725000, Shaanxi Province, China
| | - Tao Li
- Department of Infectious Diseases, Ankang Central Hospital, 85 South Jinzhou Road, Ankang, 725000, Shaanxi Province, China
| | - Xuemin Dong
- Laboratory of Molecular Pathology and Tuberculosis Diseases, Ankang Central Hospital, 85 South Jinzhou Road, Ankang, 725000, Shaanxi Province, China
| |
Collapse
|
16
|
Assinger A, Schrottmaier WC, Salzmann M, Rayes J. Platelets in Sepsis: An Update on Experimental Models and Clinical Data. Front Immunol 2019; 10:1687. [PMID: 31379873 PMCID: PMC6650595 DOI: 10.3389/fimmu.2019.01687] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/04/2019] [Indexed: 12/22/2022] Open
Abstract
Beyond their important role in hemostasis, platelets play a crucial role in inflammatory diseases. This becomes apparent during sepsis, where platelet count and activation correlate with disease outcome and survival. Sepsis is caused by a dysregulated host response to infection, leading to organ dysfunction, permanent disabilities, or death. During sepsis, tissue injury results from the concomitant uncontrolled activation of the complement, coagulation, and inflammatory systems as well as platelet dysfunction. The balance between the systemic inflammatory response syndrome (SIRS) and the compensatory anti-inflammatory response (CARS) regulates sepsis outcome. Persistent thrombocytopenia is considered as an independent risk factor of mortality in sepsis, although it is still unclear whether the drop in platelet count is the cause or the consequence of sepsis severity. The role of platelets in sepsis development and progression was addressed in different experimental in vivo models, particularly in mice, that represent various aspects of human sepsis. The immunomodulatory function of platelets depends on the experimental model, time, and type of infection. Understanding the molecular mechanism of platelet regulation in inflammation could bring us one step closer to understand this important aspect of primary hemostasis which drives thrombotic as well as bleeding complications in patients with sterile and infectious inflammation. In this review, we summarize the current understanding of the contribution of platelets to sepsis severity and outcome. We highlight the differences between platelet receptors in mice and humans and discuss the potential and limitations of animal models to study platelet-related functions in sepsis.
Collapse
Affiliation(s)
- Alice Assinger
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | - Manuel Salzmann
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Julie Rayes
- Institute of Cardiovascular Science, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
17
|
Kang XQ, Shu GF, Jiang SP, Xu XL, Qi J, Jin FY, Liu D, Xiao YH, Lu XY, Du YZ. Effective targeted therapy for drug-resistant infection by ICAM-1 antibody-conjugated TPGS modified β-Ga 2O 3:Cr 3+ nanoparticles. Theranostics 2019; 9:2739-2753. [PMID: 31244919 PMCID: PMC6568169 DOI: 10.7150/thno.33452] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/18/2019] [Indexed: 12/02/2022] Open
Abstract
The prevalence of antibiotic resistance and lack of alternative drugs have posed an increasing threat to public health. Here, we prepared β-Ga2O3:Cr3+ nanoparticles modified with ICAM1-antibody-conjugated TPGS (I-TPGS/Ga2O3) as a novel antibiotic carrier for the treatment of drug-resistant infections. Methods: I-TPGS/Ga2O3 were firstly characterized by measuring particle size, morphology, crystal structure, drug loading capacity, and in vitro drug release behaviors. The in vitro antibacterial activities of I-TPGS/Ga2O3/TIG were evaluated using standard and drug-resistant bacteria. The internalization of I-TPGS/Ga2O3 was observed by fluorescence confocal imaging, and the expression levels of the efflux pump genes of TRKP were analyzed by real-time RT-PCR. In vitro cellular uptake and in vivo biodistribution study were performed to investigate the targeting specificity of I-TPGS/Ga2O3 using HUEVC and acute pneumonia mice, respectively. The in vivo anti-infective efficacy and biosafety of I-TPGS/Ga2O3/TIG were finally evaluated using acute pneumonia mice. Results: It was found that TPGS could down-regulate the over-expression of the efflux pump genes, thus decreasing the efflux pump activity of bacteria. I-TPGS/Ga2O3 with small particle size and uniform distribution facilitated their internalization in bacteria, and the TPGS modification resulted in a significant reduction in the efflux of loaded antibiotics. These properties rendered the encapsulated tigecycline to exert a stronger antibacterial activity both in vitro and in vivo. Additionally, targeted delivery of I-TPGS/Ga2O3 mediated by ICAM1 antibodies contributed to a safe and effective therapy. Conclusion: It is of great value to apply I-TPGS/Ga2O3 as a novel and effective antibiotic delivery system for the treatment of drug-resistant infections.
Collapse
|
18
|
Understanding Platelets in Infectious and Allergic Lung Diseases. Int J Mol Sci 2019; 20:ijms20071730. [PMID: 30965568 PMCID: PMC6480134 DOI: 10.3390/ijms20071730] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/02/2019] [Accepted: 04/05/2019] [Indexed: 12/29/2022] Open
Abstract
Emerging evidence suggests that platelets, cytoplasmic fragments derived from megakaryocytes, can no longer be considered just as mediators in hemostasis and coagulation processes, but as key modulators of immunity. Platelets have received increasing attention as the emergence of new methodologies has allowed the characterization of their components and functions in the immune continuum. Platelet activation in infectious and allergic lung diseases has been well documented and associated with bacterial infections reproduced in several animal models of pulmonary bacterial infections. Direct interactions between platelets and bacteria have been associated with increased pulmonary platelet accumulation, whereas bacterial-derived toxins have also been reported to modulate platelet function. Recently, platelets have been found extravascular in the lungs of patients with asthma, and in animal models of allergic lung inflammation. Their ability to interact with immune and endothelial cells and secrete immune mediators makes them one attractive target for biomarker identification that will help characterize their contribution to lung diseases. Here, we present an original review of the last advances in the platelet field with a focus on the contribution of platelets to respiratory infections and allergic-mediated diseases.
Collapse
|
19
|
Rondina MT, Zimmerman GA. The Role of Platelets in Inflammation. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00028-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
|
20
|
The Role of Platelets in Antimicrobial Host Defense. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00029-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
|
21
|
Claushuis TAM, de Vos AF, Roelofs JJTH, de Boer OJ, van 't Veer C, van der Poll T. Platelet-Dense Granules Worsen Pre-Infection Thrombocytopenia during Gram-Negative Pneumonia-Derived Sepsis. J Innate Immun 2018; 11:168-180. [PMID: 30557883 DOI: 10.1159/000494147] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 09/23/2018] [Indexed: 12/12/2022] Open
Abstract
Platelet-dense (δ) granules are important for platelet function. Platelets contribute to host defense and vascular integrity during pneumonia and sepsis, and δ granule products, including adenosine diphosphate (ADP), can influence inflammatory responses. We therefore aimed to study the role of platelet δ granules in the host response during sepsis. Hermansky-Pudlak syndrome (Hps)3coa mice (with reduced δ granule content), mice treated with the platelet ADP receptor inhibitor clopidogrel, and appropriate control mice were infected with the human sepsis pathogen Klebsiella pneumoniae via the airways to induce pneumonia and sepsis. In order to override potential redundancy in platelet functions, we also studied Hps3coa and control mice with moderate antibody-induced thrombocytopenia (10%) prior to infection. We found that sepsis-induced thrombocytopenia tended to be less severe in Hps3coa mice, and was significantly ameliorated in Hps3coa mice with low pre-infection platelet counts. Bacterial growth was similar in Hps3coa and control mice in the presence of normal platelet counts prior to infection, but lower in the lungs of Hps3coa mice with low pre-infection platelet counts. Hps3coa mice had unaltered lung pathology and distant organ injury during pneumosepsis, irrespective of pre-infection platelet counts; lung bleeding did not differ between Hps3coa and control mice. Clopidogrel did not influence any host response parameter. These data suggest that platelet δ granules can play a detrimental role in pneumosepsis by aggravating thrombocytopenia and impairing local antibacterial defense, but that these unfavorable effects only become apparent in the presence of low platelet counts.
Collapse
Affiliation(s)
- Theodora A M Claushuis
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands,
| | - Alex F de Vos
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Onno J de Boer
- Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Cornelis van 't Veer
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Abstract
Pneumonia is a type of acute lower respiratory infection that is common and severe. The outcome of lower respiratory infection is determined by the degrees to which immunity is protective and inflammation is damaging. Intercellular and interorgan signaling networks coordinate these actions to fight infection and protect the tissue. Cells residing in the lung initiate and steer these responses, with additional immunity effectors recruited from the bloodstream. Responses of extrapulmonary tissues, including the liver, bone marrow, and others, are essential to resistance and resilience. Responses in the lung and extrapulmonary organs can also be counterproductive and drive acute and chronic comorbidities after respiratory infection. This review discusses cell-specific and organ-specific roles in the integrated physiological response to acute lung infection, and the mechanisms by which intercellular and interorgan signaling contribute to host defense and healthy respiratory physiology or to acute lung injury, chronic pulmonary disease, and adverse extrapulmonary sequelae. Pneumonia should no longer be perceived as simply an acute infection of the lung. Pneumonia susceptibility reflects ongoing and poorly understood chronic conditions, and pneumonia results in diverse and often persistent deleterious consequences for multiple physiological systems.
Collapse
Affiliation(s)
- Lee J Quinton
- Pulmonary Center, Boston University School of Medicine , Boston, Massachusetts
| | - Allan J Walkey
- Pulmonary Center, Boston University School of Medicine , Boston, Massachusetts
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University School of Medicine , Boston, Massachusetts
| |
Collapse
|
23
|
Lee CH, Chuah SK, Tai WC, Chen IL. Platelet reactivity in diabetic patients with invasive Klebsiella pneumoniae liver abscess syndrome. Infect Drug Resist 2018; 11:1669-1676. [PMID: 30349324 PMCID: PMC6183545 DOI: 10.2147/idr.s174913] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Objective Platelets catalyze the development of hyperinflammation and microthrombosis and contribute to increases in accumulation of circulating platelet-leukocyte complex, the key event in the development of disseminated infection. Subjects and methods To determine the relationships of platelet activity in diabetic patients with invasive Klebsiella pneumoniae liver abscess syndrome (IKLAS), a total of 175 diabetic patients with community-acquired Klebsiella pneumoniae (KP) bacteremia were included in this study. We compared the platelet reactivity of 40 patients with IKLAS, 40 patients with non-IKLAS, and eight healthy controls using a whole-blood flow cytometry-based assay. Results Patients who were infected with strains expressing K1/K2 serotype (adjusted odds ratio [AOR], 8.81; 95% CI, 2.18–35.53) and those with HbA1c ≥9% (AOR, 4.97; 95% CI, 1.73–14.23) were more likely to present with IKLAS, whereas those who had recent therapy with aspirin (AOR, 0.17; 95% CI, 0.04–0.79) were less likely to present with IKLAS. Among patients with IKLAS, patients with a poor glycemic control were more likely to present with hepatic venous thrombo-phlebitis than those with suboptimal or good glycemic control (P=0.03). Patients with IKLAS had a higher median fluorescence intensity of the platelet membrane expression of P-selectin than those with non-IKLAS (78.0 vs 28.0, P<0.001) and controls (78.0 vs 22.0, P< 0.001). The IKLAS group also demonstrated a significantly higher platelet-monocyte aggregation and higher plasma levels of PF-4 than the non-IKLAS group (47.0 vs 18.0 and 47.0 vs 4.0, respectively, both P <0.001) and controls (46.0 vs 24.0 and 46.0 vs 13.0, respectively, both P <0.001). Conclusion Diabetic patients with IKLAS demonstrated platelet hyperreactivity, which may be associated with a higher risk for vascular complications.
Collapse
Affiliation(s)
- Chen-Hsiang Lee
- Department of Internal Medicine, Division of Infectious Diseases, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan, .,Chang Gung University College of Medicine, Kaohsiung, Taiwan,
| | - Seng-Kee Chuah
- Chang Gung University College of Medicine, Kaohsiung, Taiwan, .,Department of Internal Medicine, Division of Gastroenterology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Wei-Chen Tai
- Chang Gung University College of Medicine, Kaohsiung, Taiwan, .,Department of Internal Medicine, Division of Gastroenterology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - I-Ling Chen
- Department of Pharmacology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| |
Collapse
|
24
|
Claushuis TAM, de Stoppelaar SF, de Vos AF, Grootemaat AE, van der Wel NN, Roelofs JJTH, Ware J, Van't Veer C, van der Poll T. Nbeal2 Deficiency Increases Organ Damage but Does Not Affect Host Defense During Gram-Negative Pneumonia-Derived Sepsis. Arterioscler Thromb Vasc Biol 2018; 38:1772-1784. [PMID: 29930006 DOI: 10.1161/atvbaha.118.311332] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective- Nbeal2-/- mice, a model of human gray platelet syndrome, have reduced neutrophil granularity and impaired host defense against systemic Staphylococcus aureus infection. We here aimed to study the role of Nbeal2 deficiency in both leukocytes and platelets during gram-negative pneumonia and sepsis. Approach and Results- We studied the role of Nbeal2 in platelets and leukocytes during murine pneumonia and sepsis by Klebsiella pneumoniae. Apart from platelet α-granule deficiency and reduced neutrophil granularity, also monocyte granularity was reduced in Nbeal2-/- mice, whereas plasma levels of MPO (myeloperoxidase), elastase, NGAL (neutrophil gelatinase-associated lipocalin), and MMP-9 (matrix metalloproteinase 9), and leukocyte CD11b expression were increased. Nbeal2-/- leukocytes showed unaltered in vitro antibacterial response and phagocytosis capacity against Klebsiella, and unchanged reactive nitrogen species and cytokine production. Also during Klebsiella pneumonia and sepsis, Nbeal2-/- mice had similar bacterial growth in lung and distant body sites, with enhanced leukocyte migration to the bronchoalveolar space. Despite similar infection-induced inflammation, organ damage was increased in Nbeal2-/- mice, which was also seen during endotoxemia. Platelet-specific Nbeal2 deficiency did not influence leukocyte functions, indicating that Nbeal2 directly modifies leukocytes. Transfusion of Nbeal2-/- but not of Nbeal2+/+ platelets into thrombocytopenic mice was associated with bleeding in the lung but similar host defense, pointing at a role for platelet α-granules in maintaining vascular integrity but not host defense during Klebsiella pneumosepsis. Conclusions- These data show that Nbeal2 deficiency-resulting in gray platelet syndrome-affects platelets, neutrophils, and monocytes, with intact host defense but increased organ damage during gram-negative pneumosepsis.
Collapse
Affiliation(s)
- Theodora A M Claushuis
- From the Center for Experimental and Molecular Medicine (T.A.M.C., S.F.d.S., A.F.d.V., C.v.V., T.v.d.P.)
| | - Sacha F de Stoppelaar
- From the Center for Experimental and Molecular Medicine (T.A.M.C., S.F.d.S., A.F.d.V., C.v.V., T.v.d.P.)
| | - Alex F de Vos
- From the Center for Experimental and Molecular Medicine (T.A.M.C., S.F.d.S., A.F.d.V., C.v.V., T.v.d.P.)
| | - Anita E Grootemaat
- Academic Medical Center, University of Amsterdam, The Netherlands; Electron Microscopy Center Amsterdam, Medical Biology, Academic Medical Center, The Netherlands (A.E.G., N.N.v.d.W.)
| | - Nicole N van der Wel
- Academic Medical Center, University of Amsterdam, The Netherlands; Electron Microscopy Center Amsterdam, Medical Biology, Academic Medical Center, The Netherlands (A.E.G., N.N.v.d.W.)
| | | | - Jerry Ware
- University of Arkansas for Medical Sciences, Little Rock (J.W.)
| | - Cornelis Van't Veer
- From the Center for Experimental and Molecular Medicine (T.A.M.C., S.F.d.S., A.F.d.V., C.v.V., T.v.d.P.)
| | - Tom van der Poll
- From the Center for Experimental and Molecular Medicine (T.A.M.C., S.F.d.S., A.F.d.V., C.v.V., T.v.d.P.)
- Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, The Netherlands (T.v.d.P.)
| |
Collapse
|
25
|
Amison RT, O'Shaughnessy BG, Arnold S, Cleary SJ, Nandi M, Pitchford SC, Bragonzi A, Page CP. Platelet Depletion Impairs Host Defense to Pulmonary Infection with Pseudomonas aeruginosa in Mice. Am J Respir Cell Mol Biol 2018; 58:331-340. [PMID: 28957635 DOI: 10.1165/rcmb.2017-0083oc] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Platelets have been implicated in pulmonary inflammatory cell recruitment after exposure to allergic and nonallergic stimuli, but little is known about the role of platelets in response to pulmonary infection with Pseudomonas aeruginosa. In this study, we have investigated the impact of the experimental depletion of circulating platelets on a range of inflammatory and bacterial parameters, and their subsequent impact on mortality in a murine model of pulmonary infection with P. aeruginosa. P. aeruginosa infection in mice induced a mild, but significant, state of peripheral thrombocytopenia in addition to pulmonary platelet accumulation. Increased platelet activation was detected in infected mice through increased levels of the platelet-derived mediators, platelet factor-4 and β-thromboglobulin, in BAL fluid and blood plasma. In mice depleted of circulating platelets, pulmonary neutrophil recruitment was significantly reduced 24 hours after infection, whereas the incidence of systemic dissemination of bacteria was significantly increased compared with non-platelet-depleted control mice. Furthermore, mortality rates were increased in bacterial-infected mice depleted of circulating platelets. This work demonstrates a role for platelets in the host response toward a gram-negative bacterial respiratory infection.
Collapse
Affiliation(s)
- Richard T Amison
- 1 Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| | - Blaze G O'Shaughnessy
- 1 Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| | - Stephanie Arnold
- 1 Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| | - Simon J Cleary
- 1 Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| | - Manasi Nandi
- 2 British Heart Foundation Centre for Cardiovascular Research, King's College London, London, United Kingdom; and
| | - Simon C Pitchford
- 1 Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| | - Alessandra Bragonzi
- 3 Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation, and Infectious Diseases, Scientific Institute for Research, Hospitalisation and Health Care San Raffaele Scientific Institute, Milan, Italy
| | - Clive P Page
- 1 Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science and
| |
Collapse
|
26
|
Middleton EA, Rondina MT, Schwertz H, Zimmerman GA. Amicus or Adversary Revisited: Platelets in Acute Lung Injury and Acute Respiratory Distress Syndrome. Am J Respir Cell Mol Biol 2018; 59:18-35. [PMID: 29553813 PMCID: PMC6039872 DOI: 10.1165/rcmb.2017-0420tr] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 02/20/2018] [Indexed: 12/21/2022] Open
Abstract
Platelets are essential cellular effectors of hemostasis and contribute to disease as circulating effectors of pathologic thrombosis. These are their most widely known biologic activities. Nevertheless, recent observations demonstrate that platelets have a much more intricate repertoire beyond these traditional functions and that they are specialized for contributions to vascular barrier integrity, organ repair, antimicrobial host defense, inflammation, and activities across the immune continuum. Paradoxically, on the basis of clinical investigations and animal models of disease, some of these newly discovered activities of platelets appear to contribute to tissue injury. Studies in the last decade indicate unique interactions of platelets and their precursor, the megakaryocyte, in the lung and implicate platelets as essential effectors in experimental acute lung injury and clinical acute respiratory distress syndrome. Additional discoveries derived from evolving work will be required to precisely define the contributions of platelets to complex subphenotypes of acute lung injury and to determine if these remarkable and versatile blood cells are therapeutic targets in acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Elizabeth A. Middleton
- Division of Pulmonary and Critical Care Medicine, and
- Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - Matthew T. Rondina
- Division of General Internal Medicine, Department of Internal Medicine
- Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - Hansjorg Schwertz
- Division of Vascular Surgery, Department of Surgery, and
- Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - Guy A. Zimmerman
- Division of Pulmonary and Critical Care Medicine, and
- Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
27
|
Finsterbusch M, Schrottmaier WC, Kral-Pointner JB, Salzmann M, Assinger A. Measuring and interpreting platelet-leukocyte aggregates. Platelets 2018; 29:677-685. [PMID: 29461910 PMCID: PMC6178087 DOI: 10.1080/09537104.2018.1430358] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Platelets, besides their specialised role in haemostasis and atherothrombosis, actively modulate innate and adaptive immune responses with crucial roles in immune surveillance, inflammation and host defence during infection. An important prerequisite for platelet-mediated changes of immune functions involves direct engagement with different types of leukocytes. Indeed, increased platelet-leukocyte aggregates (PLAs) within the circulation and/or locally at the site of inflammation represent markers of many thrombo-inflammatory diseases, such as cardiovascular diseases, acute lung injury, renal and cerebral inflammation. Therefore, measurement of PLAs could provide an attractive and easily accessible prognostic and/or diagnostic tool for many diseases. To measure PLAs in different (patho-)physiological settings in human and animal models flow cytometric and microscopic approaches have been applied. These techniques represent complementary tools to study different aspects relating to the involvement of leukocyte subtypes and molecules, as well as location of PLAs within tissues, dynamics of their interactions and/or dynamic changes in leukocyte and platelet behaviour. This review summarises various approaches to measure and interpret PLAs and discusses potential experimental factors influencing platelet binding to leukocytes. Furthermore, we summarise insights gained from studies regarding the underlying mechanism of platelet-leukocyte interactions and discuss implications of these interactions in health and disease.
Collapse
Affiliation(s)
- Michaela Finsterbusch
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Waltraud C Schrottmaier
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Julia B Kral-Pointner
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Manuel Salzmann
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| | - Alice Assinger
- a Department for Vascular Biology and Thrombosis Research , Centre for Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria
| |
Collapse
|
28
|
Platelet glycoprotein VI aids in local immunity during pneumonia-derived sepsis caused by gram-negative bacteria. Blood 2017; 131:864-876. [PMID: 29187378 DOI: 10.1182/blood-2017-06-788067] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 11/25/2017] [Indexed: 12/13/2022] Open
Abstract
Platelet collagen receptor glycoprotein VI (GPVI) and podoplanin receptor C-type lectin-like receptor 2 (CLEC2) are receptors implicated in platelet activation that both signal via an immunoreceptor tyrosine-based activation motif. Platelets are necessary for host defense and prevention of hemorrhage during sepsis, but the role of platelet GPVI and CLEC2 herein is unknown. To investigate this, we infected mice depleted of platelet GPVI or CLEC2 by antibody treatment or GPVI-/- mice with the common human sepsis pathogen Klebsiella pneumoniae via the airways to induce pneumonia-derived sepsis. The GPVI ligand collagen and the CLEC2 ligand podoplanin were constitutively present in the lung, whereas the GPVI ligands fibrin and histone were induced during pneumonia. During late-stage infection, both mice depleted of GPVI and GPVI-/- mice showed increased bacterial growth in lungs, and GPVI-/- mice also showed increased bacterial growth in distant body sites. Despite higher bacterial loads, GPVI-depleted mice showed reduced platelet numbers, platelet activation, and platelet-leukocyte complex formation in the bronchoalveolar space. Consistently, in human whole blood, GPVI stimulation of platelets increased platelet-leukocyte complex formation and leukocyte activation, which was accompanied by enhanced phagocytosis of Klebsiella GPVI-depleted mice showed increased lung hemorrhage during infection, but not to the extent observed in platelet-depleted mice, and lung bleeding was not significantly different between GPVI-/- and wild-type mice. CLEC2 depletion did not affect any of the responses during pneumonia. These results suggest that platelet GPVI, but not CLEC2, contributes to local host defense during pneumonia-derived sepsis by enhancing leukocyte function.
Collapse
|
29
|
Ding C, van 't Veer C, Roelofs JJTH, Shukla M, McCrae KR, Revenko AS, Crosby J, van der Poll T. Limited role of kininogen in the host response during gram-negative pneumonia-derived sepsis. Am J Physiol Lung Cell Mol Physiol 2017; 314:L397-L405. [PMID: 29122754 DOI: 10.1152/ajplung.00288.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
High-molecular-weight kininogen (HK), together with factor XI, factor XII and prekallikrein, is part of the contact system that has proinflammatory, prothrombotic, and vasoactive properties. We hypothesized that HK plays a role in the host response during pneumonia-derived sepsis. To this end mice were depleted of kininogen (KNG) to plasma HK levels of 28% of normal by repeated treatment with a specific antisense oligonucleotide (KNG ASO) for 3 wk before infection with the common human sepsis pathogen Klebsiella pneumoniae via the airways. Whereas plasma HK levels increased during infection in mice treated with a scrambled control ASO (Ctrl ASO), HK level in the KNG ASO-treated group remained reduced to 25-30% of that in the corresponding Ctrl ASO group both before and after infection. KNG depletion did not influence bacterial growth in lungs or dissemination to distant body sites. KNG depletion was associated with lower lung CXC chemokine and myeloperoxidase levels but did not impact neutrophil influx, lung pathology, activation of the vascular endothelium, activation of the coagulation system, or the extent of distant organ injury. These results were corroborated by studies in mice with a genetic deficiency of KNG, which were indistinguishable from wild-type mice during Klebsiella-induced sepsis. Both KNG depletion and KNG deficiency were associated with strongly reduced plasma prekallikrein levels, indicating the carrier function of HK for this zymogen. This study suggests that KNG does not significantly contribute to the host defense during gram-negative pneumonia-derived sepsis.
Collapse
Affiliation(s)
- Chao Ding
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University , Nanjing , China.,Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands
| | - Cornelis van 't Veer
- Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam , The Netherlands
| | - Meenal Shukla
- Departments of Hematology-Oncology and Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Keith R McCrae
- Departments of Hematology-Oncology and Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Alexey S Revenko
- Antisense Drug Discovery, IONIS Pharmaceuticals, Carlsbad, California
| | - Jeff Crosby
- Antisense Drug Discovery, IONIS Pharmaceuticals, Carlsbad, California
| | - Tom van der Poll
- Center of Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam , Amsterdam , The Netherlands.,Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam , The Netherlands
| |
Collapse
|
30
|
Amison R, Arnold S, O'Shaughnessy B, Cleary S, Ofoedu J, Idzko M, Page C, Pitchford S. Lipopolysaccharide (LPS) induced pulmonary neutrophil recruitment and platelet activation is mediated via the P2Y1 and P2Y14 receptors in mice. Pulm Pharmacol Ther 2017; 45:62-68. [DOI: 10.1016/j.pupt.2017.05.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/04/2017] [Accepted: 05/06/2017] [Indexed: 11/29/2022]
|
31
|
Claushuis TAM, de Stoppelaar SF, Stroo I, Roelofs JJTH, Ottenhoff R, van der Poll T, Van't Veer C. Thrombin contributes to protective immunity in pneumonia-derived sepsis via fibrin polymerization and platelet-neutrophil interactions. J Thromb Haemost 2017; 15:744-757. [PMID: 28092405 DOI: 10.1111/jth.13625] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Indexed: 01/20/2023]
Abstract
Essentials Immunity and coagulation are linked during sepsis but the role of thrombin is not fully elucidated. We investigated the effect of thrombin inhibition on murine Klebsiella pneumosepsis outcome. Thrombin is crucial for survival and limiting bacterial growth in pneumonia derived sepsis. Thrombin improves host defense via fibrin and enhancement of platelet-neutrophil interactions. SUMMARY Background Innate immunity and coagulation are closely linked during sepsis. Their interaction can be detrimental to the outcome because of microvascular failure but can also enhance host defense. The role of thrombin therein has not been fully elucidated. Objective We aimed to investigate the contribution of thrombin to the host response during pneumonia-derived sepsis. Methods Mice treated with the specific thrombin inhibitor dabigatran or control chow were infected with the common human sepsis pathogen Klebsiella (K.) pneumoniae via the airways. In subsequent infection experiments, mice were additionally treated with ancrod to deplete fibrinogen. Ex vivo Klebsiella growth was assessed by incubating human whole blood or specific blood components in various conditions with Klebsiella. Results Thrombin inhibition by dabigatran enhanced bacterial outgrowth and spreading, and accelerated mortality. Thrombin inhibition did not influence neutrophil recruitment to the lung or activation or neutrophil extracellular trap formation. Dabigatran reduced D-dimer formation and fibrin deposition in the lung. Fibrin depletion also enhanced bacterial outgrowth and spreading, and thrombin inhibition had no additional effect. Both thrombin and fibrin polymerization inhibited ex vivo Klebsiella outgrowth in human whole blood, which was neutrophil dependent, and the effect of thrombin required the presence of platelets and platelet protease activated receptor-1. In vivo thrombin inhibition reduced platelet-neutrophil complex formation and endothelial cell activation, but did not prevent sepsis-induced thrombocytopenia or organ damage. Conclusions These results suggest that thrombin plays an important role in protective immunity during pneumonia-derived sepsis by fibrin polymerization and enhancement of platelet-neutrophil interactions.
Collapse
Affiliation(s)
- T A M Claushuis
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - S F de Stoppelaar
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - I Stroo
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Immunopathology, Sanquin Research, Amsterdam, the Netherlands
| | - J J T H Roelofs
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - R Ottenhoff
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - T van der Poll
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - C Van't Veer
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
32
|
Birnie E, Koh GCKW, Löwenberg EC, Meijers JCM, Maude RR, Day NPJ, Peacock SJ, van der Poll T, Wiersinga WJ. Increased Von Willebrand factor, decreased ADAMTS13 and thrombocytopenia in melioidosis. PLoS Negl Trop Dis 2017; 11:e0005468. [PMID: 28296884 PMCID: PMC5376340 DOI: 10.1371/journal.pntd.0005468] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 03/31/2017] [Accepted: 03/08/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Melioidosis, caused by bioterror treat agent Burkholderia pseudomallei, is an important cause of community-acquired Gram-negative sepsis in Southeast Asia and Northern Australia. New insights into the pathogenesis of melioidosis may help improve treatment and decrease mortality rates from this dreadful disease. We hypothesized that changes in Von Willebrand factor (VWF) function should occur in melioidosis, based on the presence of endothelial stimulation by endotoxin, pro-inflammatory cytokines and thrombin in melioidosis, and investigated whether this impacted on outcome. METHODS/PRINCIPAL FINDINGS We recruited 52 controls and 34 culture-confirmed melioidosis patients at Sappasithiprasong Hospital in Ubon Ratchathani, Thailand. All subjects were diabetic. Platelet counts in melioidosis patients were lower compared to controls (p = 0.0001) and correlated with mortality (p = 0.02). VWF antigen levels were higher in patients (geometric mean, 478 U/dl) compared to controls (166 U/dL, p<0.0001). The high levels of VWF in melioidosis appeared to be due to increased endothelial stimulation (VWF propeptide levels were elevated, p<0.0001) and reduced clearance (ADAMTS13 reduction, p<0.0001). However, VWF antigen levels did not correlate with platelet counts implying that thrombocytopenia in acute melioidosis has an alternative cause. CONCLUSIONS/SIGNIFICANCE Thrombocytopenia is a key feature of melioidosis and is correlated with mortality. Additionally, excess VWF and ADAMTS13 deficiency are features of acute melioidosis, but are not the primary drivers of thrombocytopenia in melioidosis. Further studies on the role of thrombocytopenia in B. pseudomallei infection are needed.
Collapse
Affiliation(s)
- Emma Birnie
- Center for Experimental and Molecular Medicine, Division of Infectious Diseases, Academic Medical Center, Amsterdam, The Netherlands
| | - Gavin C. K. W. Koh
- Center for Experimental and Molecular Medicine, Division of Infectious Diseases, Academic Medical Center, Amsterdam, The Netherlands
- Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kindom
- Department of Infection and Tropical Medicine, Heartlands Hospital, Birmingham, United Kindom
| | - Ester C. Löwenberg
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Joost C. M. Meijers
- Department of Experimental Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
- Department of Plasma Proteins, Sanquin Research, Amsterdam, The Netherlands
| | | | - Nicholas P. J. Day
- Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- The Centre for Clinical Vaccinology and Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Churchill Hospital, Oxford, United Kindom
| | - Sharon J. Peacock
- Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kindom
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Division of Infectious Diseases, Academic Medical Center, Amsterdam, The Netherlands
| | - W. Joost Wiersinga
- Center for Experimental and Molecular Medicine, Division of Infectious Diseases, Academic Medical Center, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
33
|
Abstract
Platelets are small, anucleate circulating cells that possess a dynamic repertoire of functions spanning the hemostatic, inflammatory, and immune continuum. Once thought to be merely cell fragments with responses limited primarily to acute hemostasis and vascular wall repair, platelets are now increasingly recognized as key sentinels and effector cells regulating host responses to many inflammatory and infectious cues. Platelet granules, including α-granules and dense-granules, store hundreds of factors and secrete these mediators in response to activating signals. The cargo packaged and stored within platelet granules orchestrates communication between platelets and other circulating cells, augments host defense mechanisms to invading pathogens and tumor cells, and - in some settings - drives dysregulated and injurious responses. This focused review will highlight several of the established and emerging mechanisms and roles of platelet secretion in inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Bhanu K Manne
- a The University of Utah Molecular Medicine Program , Salt Lake City , Utah , USA
| | | | - Matthew T Rondina
- a The University of Utah Molecular Medicine Program , Salt Lake City , Utah , USA.,c Department of Internal Medicine , Salt Lake City , Utah , USA.,d The GRECC, George E. Wahlen Salt Lake City VAMC , Salt Lake City , Utah , USA
| |
Collapse
|
34
|
Riffo-Vasquez Y, Somani A, Man F, Amison R, Pitchford S, Page CP. A Non-Anticoagulant Fraction of Heparin Inhibits Leukocyte Diapedesis into the Lung by an Effect on Platelets. Am J Respir Cell Mol Biol 2016; 55:554-563. [PMID: 27181499 DOI: 10.1165/rcmb.2015-0172oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
We have investigated whether the mechanism by which the non-anticoagulant N-acetyl-de-O-sulfated-heparin (NSH) inhibits leukocyte infiltration is mediated by an effect on platelet function. We show that oral treatment with two doses of NSH significantly inhibits eosinophil and neutrophil recruitment into the lungs. Intravital microscopy analysis shows that NSH inhibits leukocyte and platelet diapedesis in the microcirculation of the cremaster muscle and in the trachea. More importantly, there were significantly lower numbers of leukocytes recruited into the lung in response to LPS in thrombocytopenic mice when transfused with platelets pretreated with NSH in vitro when compared with mice transfused with untreated platelets. Using intravital analysis of the microvasculature of the cremaster muscle, we have demonstrated that the reinfusion of activated platelets significantly re-established leukocyte diapedesis in response to LPS but that this effect was not observed when platelets were pretreated in vitro with NSH. Finally, we investigated whether the effect of NSH altered the expression of adhesion molecules on the surface of platelets and leukocytes in blood samples collected from mice treated orally with NSH. Our results demonstrate that NSH significantly inhibited the detection of P-selectin as evaluated by flow cytometry, confirming that part of the antiinflammatory action of NSH is via an effect on platelet function.
Collapse
Affiliation(s)
- Yanira Riffo-Vasquez
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Alysha Somani
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Francis Man
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Richard Amison
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Simon Pitchford
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Clive P Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| |
Collapse
|
35
|
Hurley SM, Lutay N, Holmqvist B, Shannon O. The Dynamics of Platelet Activation during the Progression of Streptococcal Sepsis. PLoS One 2016; 11:e0163531. [PMID: 27656898 PMCID: PMC5033464 DOI: 10.1371/journal.pone.0163531] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 09/09/2016] [Indexed: 01/11/2023] Open
Abstract
Platelets contribute to inflammation however, the role of platelet activation during the pathophysiological response to invasive bacterial infection and sepsis is not clear. Herein, we have investigated platelet activation in a mouse model of invasive Streptococcus pyogenes infection at 5, 12, and 18 hours post infection and correlated this to parameters of infection. The platelet population in ex-vivo blood samples showed no increased integrin activation or surface presentation of CD62P, however platelet-neutrophil complex formation and plasma levels of CD62P were increased during bacterial dissemination and the progression of sepsis, indicating that platelet activation had occurred in vivo. Platelet-neutrophil complex formation was the most discriminatory marker of platelet activation. Platelet-neutrophil complexes were increased above baseline levels during early sepsis but decreased to significantly lower levels than baseline during late sepsis. The removal of these complexes from the circulation coincided with a significant increase in organ damage and the accumulation of platelets in the liver sinusoids, suggesting that platelet activation in the circulation precedes accumulation of platelets in damaged organs. The results demonstrate that monitoring platelet activation using complementary methods may provide prognostic information during the pathogenesis of invasive S. pyogenes infection.
Collapse
Affiliation(s)
- Sinead M. Hurley
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, SE- 22184 Lund, Sweden
| | | | - Bo Holmqvist
- Imagene-iT AB, Medicon Village, SE22381 Lund, Sweden
| | - Oonagh Shannon
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, SE- 22184 Lund, Sweden
| |
Collapse
|
36
|
Hechler B, Gachet C. Purinergic Receptors in Thrombosis and Inflammation. Arterioscler Thromb Vasc Biol 2015; 35:2307-15. [PMID: 26359511 DOI: 10.1161/atvbaha.115.303395] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 08/27/2015] [Indexed: 12/23/2022]
Abstract
Under various pathological conditions, including thrombosis and inflammation, extracellular nucleotide levels may increase because of both active release and passive leakage from damaged or dying cells. Once in the extracellular compartment, nucleotides interact with plasma membrane receptors belonging to the P2 purinergic family, which are expressed by virtually all circulating blood cells and in most blood vessels. In this review, we focus on the specific role of the 3 platelet P2 receptors P2Y1, P2Y12, and P2X1 in hemostasis and arterial thrombosis. Beyond platelets, these 3 receptors, along with the P2Y2, P2Y6, and P2X7 receptors, constitute the main P2 receptors mediating the proinflammatory effects of nucleotides, which play important roles in various functions of circulating blood cells and cells of the vessel wall. Each of these P2 receptor subtypes specifically contributes to chronic or acute vascular inflammation and related diseases, such as atherosclerosis, restenosis, endotoxemia, and sepsis. The potential for therapeutic targeting of these P2 receptor subtypes is also discussed.
Collapse
Affiliation(s)
- Béatrice Hechler
- From the UMR_S949, INSERM, Strasbourg, France; Etablissement Français du Sang-Alsace (EFS-Alsace), Strasbourg, France; and Université de Strasbourg, Strasbourg, France
| | - Christian Gachet
- From the UMR_S949, INSERM, Strasbourg, France; Etablissement Français du Sang-Alsace (EFS-Alsace), Strasbourg, France; and Université de Strasbourg, Strasbourg, France.
| |
Collapse
|
37
|
de Stoppelaar SF, Claushuis TAM, Jansen MPB, Hou B, Roelofs JJTH, van 't Veer C, van der Poll T. The role of platelet MyD88 in host response during gram-negative sepsis. J Thromb Haemost 2015; 13:1709-20. [PMID: 26178922 DOI: 10.1111/jth.13048] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 06/30/2015] [Indexed: 01/14/2023]
Abstract
BACKGROUND Beside their role in hemostasis, platelets serve as sentinel cells in host defense during infection. In sepsis, platelets have been implicated in both beneficial (antibacterial) and detrimental responses (thrombosis and organ damage). Toll-like receptors and their common adaptor, myeloid differentiation factor 88 (MyD88), are essential for pathogen recognition and protective immunity. Platelets express functional Toll-like receptors and MyD88, which participate in platelet responsiveness to bacterial agonists. OBJECTIVE Considering the pivotal involvement of platelets and MyD88 in the host response to bacteria, we studied the role of platelet MyD88 in gram-negative sepsis using intravenous and airway infections with the common human sepsis pathogen Klebsiella pneumoniae. METHODS Platelet-specific Myd88(-/-) mice were generated by crossing mice with a conditional Myd88 flox allele with mice expressing Cre recombinase controlled by the platelet factor 4 promoter. In a reverse approach, full Myd88(-/-) mice were transfused with wild-type platelets. RESULTS In both settings, platelet MyD88 did not impact on bacterial growth or dissemination. In addition, platelet MyD88 did not influence hallmark sepsis responses such as thrombocytopenia, coagulation or endothelial activation, or distant organ injury. Platelet MyD88 played no role in lung pathology during pneumonia-derived sepsis. CONCLUSION Despite known literature, platelet MyD88-dependent TLR signaling does not contribute to the host response during gram-negative sepsis.
Collapse
Affiliation(s)
- S F de Stoppelaar
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - T A M Claushuis
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - M P B Jansen
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - B Hou
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chaoyang District, Beijing, China
| | - J J T H Roelofs
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - C van 't Veer
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - T van der Poll
- Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|