1
|
Vizcaino Castro A, Daemen T, Oyarce C. Strategies to reprogram anti-inflammatory macrophages towards pro-inflammatory macrophages to support cancer immunotherapies. Immunol Lett 2024; 267:106864. [PMID: 38705481 DOI: 10.1016/j.imlet.2024.106864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/25/2024] [Accepted: 04/30/2024] [Indexed: 05/07/2024]
Abstract
Tumor-associated myeloid cells, including macrophages and myeloid-derived suppressor cells, can be highly prevalent in solid tumors and play a significant role in the development of the tumor. Therefore, myeloid cells are being considered potential targets for cancer immunotherapies. In this review, we focused on strategies aimed at targeting tumor-associated macrophages (TAMs). Most strategies were studied preclinically but we also included a limited number of clinical studies based on these strategies. We describe possible underlying mechanisms and discuss future challenges and prospects.
Collapse
Affiliation(s)
- Ana Vizcaino Castro
- Laboratory of Tumor Virology and Cancer Immunotherapy, Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Toos Daemen
- Laboratory of Tumor Virology and Cancer Immunotherapy, Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Cesar Oyarce
- Laboratory of Tumor Virology and Cancer Immunotherapy, Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
2
|
Sloos PH, Maas MAW, Meijers JCM, Nieuwland R, Roelofs JJTH, Juffermans NP, Kleinveld DJB. Anti-high-mobility group box-1 treatment strategies improve trauma-induced coagulopathy in a mouse model of trauma and shock. Br J Anaesth 2023; 130:687-697. [PMID: 36967283 DOI: 10.1016/j.bja.2023.01.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 01/09/2023] [Accepted: 01/30/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND Trauma-induced coagulopathy is associated with platelet dysfunction and contributes to early mortality after traumatic injury. Plasma concentrations of the damage molecule high-mobility group box-1 (HMGB-1) increase after trauma, which may contribute to platelet dysfunction. We hypothesised that inhibition of HMGB-1 with a monoclonal antibody (mAb) or with recombinant thrombomodulin (rTM) improves trauma-induced coagulopathy in a murine model of trauma and shock. METHODS Male 129S2/SvPasOrlRJ mice were anaesthetised, mechanically ventilated, and randomised into five groups: (i) ventilation control (VENT), (ii) trauma/shock (TS), (iii) TS+anti-HMGB-1 mAb (TS+AB), (iv) TS+rTM (TS+TM), and (v) TS+anti-HMGB-1 mAb+rTM (TS+COMBI). Primary outcome was rotational thromboelastometry EXTEM. Secondary outcomes included tail bleeding time, platelet count, plasma HMGB-1 concentration, and platelet activation. RESULTS Trauma and shock resulted in a hypocoagulable thromboelastometry profile, increased plasma HMGB-1, and increased platelet activation markers. TS+AB was associated with improved clot firmness after 5 min compared with TS (34 [33-37] vs 32 [29-34] mm; P=0.043). TS+COMBI was associated with decreased clot formation time (98 [92-125] vs 122 [111-148] s; P=0.018) and increased alpha angle (77 [72-78] vs 69 [64-71] degrees; P=0.003) compared with TS. TS+COMBI also reduced tail bleeding time compared with TS (P=0.007). The TS+TM and TS+COMBI groups had higher platelet counts compared with TS (P=0.044 and P=0.041, respectively). CONCLUSIONS Inhibition of HMGB-1 early after trauma in a mouse model improves clot formation and strength, preserves platelet count, and decreases bleeding time.
Collapse
Affiliation(s)
- Pieter H Sloos
- Amsterdam UMC Location University of Amsterdam, Department of Intensive Care Medicine, Amsterdam, the Netherlands; Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam, the Netherlands
| | - M Adrie W Maas
- Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam, the Netherlands
| | - Joost C M Meijers
- Amsterdam UMC Location University of Amsterdam, Department of Experimental Vascular Medicine, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands; Sanquin Research, Department of Molecular Hematology, Amsterdam, the Netherlands
| | - Rienk Nieuwland
- Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Clinical Chemistry, Amsterdam, the Netherlands; Amsterdam UMC Location University of Amsterdam, Vesicle Observation Center, Amsterdam, the Netherlands
| | - Joris J T H Roelofs
- Amsterdam UMC Location University of Amsterdam, Department of Pathology, Amsterdam, the Netherlands
| | - Nicole P Juffermans
- Amsterdam UMC Location University of Amsterdam, Department of Intensive Care Medicine, Amsterdam, the Netherlands; Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam, the Netherlands; Onze Lieve Vrouwe Gasthuis, Department of Intensive Care Medicine, Amsterdam, the Netherlands
| | - Derek J B Kleinveld
- Amsterdam UMC Location University of Amsterdam, Department of Intensive Care Medicine, Amsterdam, the Netherlands; Amsterdam UMC Location University of Amsterdam, Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam, the Netherlands; Erasmus MC, Department of Anesthesiology, Rotterdam, the Netherlands.
| |
Collapse
|
3
|
Single-cell Sequence Analysis Combined with Multiple Machine Learning to Identify Markers in Sepsis Patients: LILRA5. Inflammation 2023:10.1007/s10753-023-01803-8. [PMID: 36920635 DOI: 10.1007/s10753-023-01803-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/16/2023]
Abstract
Sepsis is a disease with a very high mortality rate, mainly involving an immune-dysregulated response due to bacterial infection. Most studies are currently limited to the whole blood transcriptome level; however, at the single cell level, there is still a great deal unknown about specific cell subsets and disease markers. We obtained 29 peripheral blood single-cell sequencing data, including 66,283 cells from 10 confirmed samples of sepsis infection and 19 healthy samples. Cells related to the sepsis phenotype were identified and characterized by the "scissor" method. The regulatory relationships of sepsis-related phenotype cells in the cellular communication network were clarified using the "cell chat" method. The least absolute shrinkage and selection operator (LASSO), support vector machine (SVM), and random forest (RF) were used to identify sepsis signature genes of diagnostic value. External validation was performed using multiple datasets from the GEO database (GSE28750, GSE185263, GSE57065) and 40 clinical samples. Bayesian algorithm was used to calculate the regulatory network of LILRA5 co-expressed genes. The stability of atenolol-targeting LILRA5 was determined by molecular docking techniques. Ultimately, action trajectory and survival analyses demonstrate the effectiveness of atenolol-targeted LILRA5 in treating patients with sepsis. We successfully identified 1215 healthy phenotypic cells and 462 sepsis phenotypic cells. We focused on 447 monocytes of the sepsis phenotype. Among the cellular communications, there were a large number of differences between these cells and other immune cells showing a significant inflammatory phenotype compared to the healthy phenotypic cells. Together, the three machine learning algorithms identified the LILRA5 marker gene in sepsis patients, and validation results from multiple external datasets as well as real-world clinical samples demonstrated the robust diagnostic performance of LILRA5. The AUC values of LILRA5 in the external datasets GSE28750, GSE185263, and GSE57065 could reach 0.875, 0.940, and 0.980, in that order. Bayesian networks identified a large number of unknown regulatory relationships for LILRA5 co-expression. Molecular docking results demonstrated the possibility of atenolol targeting LILRA5 for the treatment of sepsis. Behavioral trajectory analysis and survival analysis demonstrate that atenolol has a desirable therapeutic effect. LILRA5 is a marker gene in sepsis patients, and atenolol can stably target LILRA5.
Collapse
|
4
|
Yang F, Zong H, Li F, Luo S, Zhang X, Xu Y, Zhang X. Eltrombopag modulates the phenotypic evolution and potential immunomodulatory roles of monocytes/macrophages in immune thrombocytopenia. Platelets 2022; 34:2135694. [DOI: 10.1080/09537104.2022.2135694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Affiliation(s)
- Feifei Yang
- Nanjing First Hospital, Nanjing Medical University, Nanjing, Chinaand
| | - Hui Zong
- Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Feng Li
- Nanjing First Hospital, Nanjing Medical University, Nanjing, Chinaand
| | - Shulin Luo
- Nanjing First Hospital, Nanjing Medical University, Nanjing, Chinaand
| | - Xiuqun Zhang
- Nanjing First Hospital, Nanjing Medical University, Nanjing, Chinaand
| | - Yanli Xu
- Nanjing First Hospital, Nanjing Medical University, Nanjing, Chinaand
| | - Xuezhong Zhang
- Nanjing First Hospital, Nanjing Medical University, Nanjing, Chinaand
| |
Collapse
|
5
|
Pirnie R, Gillespie KP, Weng L, Mesaros C, Blair IA. Characterization and Quantification of Oxidized High Mobility Group Box 1 Proteoforms Secreted from Hepatocytes by Toxic Levels of Acetaminophen. Chem Res Toxicol 2022; 35:1893-1902. [PMID: 35922039 PMCID: PMC9580022 DOI: 10.1021/acs.chemrestox.2c00161] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The high mobility group box 1 (HMGB1), which is released during acute acetaminophen (APAP) overdose, is thought to mediate a subsequent immune response, particularly hepatic infiltration of macrophages. The redox behavior of HMGB1 and the proteoforms of HMGB1 present in oxidative environments has been the subject of a number of confusing and contradictory studies. Therefore, a stable isotope dilution two-dimensional nanoultrahigh-performance liquid chromatography parallel reaction monitoring/high-resolution mass spectrometry method was developed in order to characterize and quantify oxidative modifications to the cysteine (Cys) residues (Cys-23, Cys-45, and Cys-106) that are present in HMGB1. Disulfide linkages were determined using carbamidoethyl derivatization before and after reduction as well as by direct analysis of disulfide cross-linked peptides. A stable isotope labeled form of HMGB1 was used as an internal standard to correct for sample to sample differences in immunoaffinity precipitation, derivatization, and electrospray ionization. Four discrete HMGB1 proteoforms were found to be released from a hepatocarcinoma cell model of APAP overdose after 24 h. Fully reduced HMGB1 with all three Cys-residues in their free thiol state accounted for 18% of the secreted HMGB1. The proteoform with disulfide between Cys-23 and Cys-45 accounted for 24% of the HMGB1. No evidence was obtained for a disulfide cross-link between Cys-106 and the other two Cys-residues. However, 45% of the HMGB1 formed a cross-link with unidentified intracellular proteins via an intermolecular disulfide bond, and 12% was present as the terminally oxidized cysteic acid. Surprisingly, there was no evidence for the formation of HMGB1 disulfides with GSH or other low molecular weight thiols. Secreted plasma HMGB1 Cys-23/Cys45 disulfide proteoform together with the Cys-106/protein disulfide proteoforms could potentially serve as early biomarkers of hepatoxicity after APAP overdose as well as biomarkers of drug-induced liver injury.
Collapse
|
6
|
Choudhuri S, Garg NJ. Platelets, Macrophages, and Thromboinflammation in Chagas Disease. J Inflamm Res 2022; 15:5689-5706. [PMID: 36217453 PMCID: PMC9547606 DOI: 10.2147/jir.s380896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/24/2022] [Indexed: 11/23/2022] Open
Abstract
Chagas disease (CD) is a major health problem in the Americas and an emerging health problem in Europe and other nonendemic countries. Several studies have documented persistence of the protozoan parasite Trypanosoma cruzi, and oxidative and inflammatory stress are major pathogenic factor. Mural and cardiac thrombi, cardiac arrhythmias, and cardiomyopathy are major clinical features of CD. During T. cruzi infection, parasite-released factors induce endothelial dysfunction along with platelet (PLT) and immune-cell activation. PLTs have a fundamental role in maintaining hemostasis and preventing bleeding after vascular injury. Excessive activation of PLTs and coagulation cascade can result in thrombosis and thromboembolic events, which are recognized to occur in seropositive individuals in early stages of CD when clinically symptomatic heart disease is not apparent. Several host and parasite factors have been identified to signal hypercoagulability and increase the risk of ischemic stroke in early phases of CD. Further, PLT interaction with immune cells and their role in host defense against pathogens and inflammatory processes have only recently been recognized and evolving. In the context of parasitic diseases, PLTs function in directly responding to T. cruzi infection, and PLT interactions with immune cells in shaping the proinflammatory or immunoregulatory function of monocytes, macrophages, and neutrophils remains elusive. How T. cruzi infection alters systemic microenvironment conditions to influence PLT and immune-cell interactions is not understood. In this review, we discuss the current literature, and extrapolate the mechanistic situations to explain how PLT and innate immune cell (especially monocytes and macrophages) interactions might be sustaining hypercoagulability and thromboinflammation in chronic CD.
Collapse
Affiliation(s)
- Subhadip Choudhuri
- Department of Microbiology and Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Nisha J Garg
- Department of Microbiology and Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
7
|
Sloos PH, Vulliamy P, van 't Veer C, Gupta AS, Neal MD, Brohi K, Juffermans NP, Kleinveld DJB. Platelet dysfunction after trauma: From mechanisms to targeted treatment. Transfusion 2022; 62 Suppl 1:S281-S300. [PMID: 35748694 PMCID: PMC9546174 DOI: 10.1111/trf.16971] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Pieter H. Sloos
- Department of Intensive Care Medicine, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Paul Vulliamy
- Centre for Trauma Sciences, Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Cornelis van 't Veer
- Center for Experimental and Molecular Medicine, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Anirban Sen Gupta
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
| | - Matthew D. Neal
- Pittsburgh Trauma and Transfusion Medicine Research Center and Division of Trauma and Acute Care SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Karim Brohi
- Centre for Trauma Sciences, Blizard Institute, Barts and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Nicole P. Juffermans
- Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Intensive Care MedicineOLVG HospitalAmsterdamThe Netherlands
| | - Derek J. B. Kleinveld
- Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Intensive Care MedicineErasmus MCRotterdamThe Netherlands
| |
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
|
Ghasemzadeh M, Ahmadi J, Hosseini E. Platelet-leukocyte crosstalk in COVID-19: How might the reciprocal links between thrombotic events and inflammatory state affect treatment strategies and disease prognosis? Thromb Res 2022; 213:179-194. [PMID: 35397313 PMCID: PMC8969450 DOI: 10.1016/j.thromres.2022.03.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/11/2022] [Accepted: 03/28/2022] [Indexed: 01/09/2023]
Abstract
Platelet-leukocyte crosstalk is commonly manifested by reciprocal links between thrombosis and inflammation. Platelet thrombus acts as a reactive matrix that recruits leukocytes to the injury site where their massive accumulation, activation and migration promote thrombotic events while triggering inflammatory responses. As a life-threatening condition with the associations between inflammation and thrombosis, COVID-19 presents diffuse alveolar damage due to exaggerated macrophage activity and cytokine storms. These events, together with direct intracellular virus invasion lead to pulmonary vascular endothelialitis, cell membranes disruption, severe endothelial injury, and thrombosis. The developing pre-alveolar thrombus provides a hyper-reactive milieu that recruits circulating leukocytes to the injury site where their activation contributes to thrombus stabilization and thrombosis propagation, primarily through the formation of Neutrophil extracellular trap (NET). NET fragments can also circulate and deposit in further distance where they may disseminate intravascular thrombosis in severe cases of disease. Thrombi may also facilitate leukocytes migration into alveoli where their accumulation and activation exacerbate cytokine storms and tissue damage, further complicating the disease. Based on these mechanisms, whether an effective anti-inflammatory protocol can prevent thrombotic events, or on the other hand; efficient antiplatelet or anticoagulant regimens may be associated with reduced cytokine storms and tissue damage, is now of interests for several ongoing researches. Thus shedding more light on platelet-leukocyte crosstalk, the review presented here discusses the detailed mechanisms by which platelets may contribute to the pathogenesis of COVID-19, especially in severe cases where their interaction with leukocytes can intensify both inflammatory state and thrombosis in a reciprocal manner.
Collapse
Affiliation(s)
- Mehran Ghasemzadeh
- Corresponding authors at: Blood Transfusion Research Centre, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Hemmat Exp. Way, Next to the Milad Tower, Tehran, Iran
| | | | - Ehteramolsadat Hosseini
- Corresponding authors at: Blood Transfusion Research Centre, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Hemmat Exp. Way, Next to the Milad Tower, Tehran, Iran
| |
Collapse
|
10
|
Wood B, Padula MP, Marks DC, Johnson L. Cryopreservation alters the immune characteristics of platelets. Transfusion 2021; 61:3432-3442. [PMID: 34636427 DOI: 10.1111/trf.16697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Cryopreserved platelets are under clinical evaluation as they offer improvements in shelf-life and potentially hemostatic effectiveness. However, the effect of cryopreservation on characteristics related to the immune function of platelets has not been examined. STUDY DESIGN AND METHODS Buffy coat derived platelets were cryopreserved at -80°C using 5%-6% dimethylsulfoxide (DMSO, n = 8). Paired testing was conducted pre-freeze (PF), post-thaw (PT0), and after 24 h of post-thaw storage at room temperature (PT24). The concentration of biological response modifiers (BRMs) in the supernatant was measured using commercial ELISAs and surface receptor abundance was assessed by flow cytometry. RESULTS Cryopreservation resulted in increased RANTES, PF4, and C3a but decreased IL-1β, OX40L, IL-13, IL-27, CD40L, and C5a concentrations in the supernatant, compared to PF samples. C4a, endocan, and HMGB1 concentrations were similar between the PF and PT0 groups. The abundance of surface-expressed P-selectin, siglec-7, TLR3, TLR7, and TLR9 was increased PT0; while CD40, CLEC2, ICAM-2, and MHC-I were decreased, compared to PF. The surface abundance of CD40L, B7-2, DC-SIGN, HCAM, TLR1, TLR2, TLR4, and TLR6 was unchanged by cryopreservation. Following 24 h of post-thaw storage, all immune associated receptors and TLRs increased to levels higher than observed on PF and PT0 platelets. CONCLUSION Cryopreservation alters the immune phenotype of platelets. Understanding the clinical implications of the observed changes in BRM release and receptor abundance are essential, as they may influence the likelihood of adverse events.
Collapse
Affiliation(s)
- Ben Wood
- Research & Development, Australian Red Cross Lifeblood, Alexandria, New South Wales, Australia.,School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Matthew P Padula
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Denese C Marks
- Research & Development, Australian Red Cross Lifeblood, Alexandria, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Lacey Johnson
- Research & Development, Australian Red Cross Lifeblood, Alexandria, New South Wales, Australia
| |
Collapse
|
11
|
Ebermeyer T, Cognasse F, Berthelot P, Mismetti P, Garraud O, Hamzeh-Cognasse H. Platelet Innate Immune Receptors and TLRs: A Double-Edged Sword. Int J Mol Sci 2021; 22:ijms22157894. [PMID: 34360659 PMCID: PMC8347377 DOI: 10.3390/ijms22157894] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 12/17/2022] Open
Abstract
Platelets are hematopoietic cells whose main function has for a long time been considered to be the maintenance of vascular integrity. They have an essential role in the hemostatic response, but they also have functional capabilities that go far beyond it. This review will provide an overview of platelet functions. Indeed, stress signals may induce platelet apoptosis through proapoptotis or hemostasis receptors, necrosis, and even autophagy. Platelets also interact with immune cells and modulate immune responses in terms of activation, maturation, recruitment and cytokine secretion. This review will also show that platelets, thanks to their wide range of innate immune receptors, and in particular toll-like receptors, and can be considered sentinels actively participating in the immuno-surveillance of the body. We will discuss the diversity of platelet responses following the engagement of these receptors as well as the signaling pathways involved. Finally, we will show that while platelets contribute significantly, via their TLRs, to immune response and inflammation, these receptors also participate in the pathophysiological processes associated with various pathogens and diseases, including cancer and atherosclerosis.
Collapse
Affiliation(s)
- Théo Ebermeyer
- INSERM U1059-SAINBIOSE, Université de Lyon, F-42023 Saint-Etienne, France; (T.E.); (F.C.); (P.M.); (O.G.)
| | - Fabrice Cognasse
- INSERM U1059-SAINBIOSE, Université de Lyon, F-42023 Saint-Etienne, France; (T.E.); (F.C.); (P.M.); (O.G.)
- Etablissement Français du Sang Auvergne-Rhône-Alpes, 25 bd Pasteur, F-42100 Saint-Étienne, France
| | - Philippe Berthelot
- Team GIMAP, CIRI—Centre International de Recherche en Infectiologie, Université de Lyon, U1111, UMR5308, F-69007 Lyon, France;
- Infectious Diseases Department, CHU de St-Etienne, F-42055 Saint-Etienne, France
| | - Patrick Mismetti
- INSERM U1059-SAINBIOSE, Université de Lyon, F-42023 Saint-Etienne, France; (T.E.); (F.C.); (P.M.); (O.G.)
- Department of Vascular Medicine and Therapeutics, INNOVTE, CHU de St-Etienne, F-42055 Saint-Etienne, France
| | - Olivier Garraud
- INSERM U1059-SAINBIOSE, Université de Lyon, F-42023 Saint-Etienne, France; (T.E.); (F.C.); (P.M.); (O.G.)
| | - Hind Hamzeh-Cognasse
- INSERM U1059-SAINBIOSE, Université de Lyon, F-42023 Saint-Etienne, France; (T.E.); (F.C.); (P.M.); (O.G.)
- Correspondence:
| |
Collapse
|
12
|
Vulliamy P, Kornblith LZ, Kutcher ME, Cohen MJ, Brohi K, Neal MD. Alterations in platelet behavior after major trauma: adaptive or maladaptive? Platelets 2021; 32:295-304. [PMID: 31986948 PMCID: PMC7382983 DOI: 10.1080/09537104.2020.1718633] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/01/2020] [Accepted: 01/14/2020] [Indexed: 12/14/2022]
Abstract
Platelets are damage sentinels of the intravascular compartment, initiating and coordinating the primary response to tissue injury. Severe trauma and hemorrhage induce profound alterations in platelet behavior. During the acute post-injury phase, platelets develop a state of impaired ex vivo agonist responsiveness independent of platelet count, associated with systemic coagulopathy and mortality risk. In patients surviving the initial insult, platelets become hyper-responsive, associated with increased risk of thrombotic events. Beyond coagulation, platelets constitute part of a sterile inflammatory response to injury: both directly through release of immunomodulatory molecules, and indirectly through modifying behavior of innate leukocytes. Both procoagulant and proinflammatory aspects have implications for secondary organ injury and multiple-organ dysfunction syndromes. This review details our current understanding of adaptive and maladaptive alterations in platelet biology induced by severe trauma, mechanisms underlying these alterations, potential platelet-focused therapies, and existing knowledge gaps and their research implications.
Collapse
Affiliation(s)
- Paul Vulliamy
- Centre for Trauma Sciences, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, E1 2AT, United Kingdom
| | - Lucy Z. Kornblith
- Department of Surgery, Zuckerberg San Francisco General Hospital and the University of California, San Francisco, San Francisco, California
| | - Matthew E. Kutcher
- Division of Trauma, Critical Care, and Acute Care Surgery, University of Mississippi Medical Center, Jackson, Mississippi
| | - Mitchell J. Cohen
- Department of Surgery, University of Colorado, Aurora, Colorado
- Ernest E Moore Shock Trauma Center at Denver Health, Denver, Colorado
| | - Karim Brohi
- Centre for Trauma Sciences, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, E1 2AT, United Kingdom
| | - Matthew D. Neal
- Division of Trauma and Acute Care Surgery, Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
13
|
Zamora C, Cantó E, Vidal S. The Dual Role of Platelets in the Cardiovascular Risk of Chronic Inflammation. Front Immunol 2021; 12:625181. [PMID: 33868242 PMCID: PMC8046936 DOI: 10.3389/fimmu.2021.625181] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/15/2021] [Indexed: 11/25/2022] Open
Abstract
Patients with chronic inflammatory diseases often exhibit cardiovascular risk. This risk is associated with the systemic inflammation that persists in these patients, causing a sustained endothelial activation. Different mechanisms have been considered responsible for this systemic inflammation, among which activated platelets have been regarded as a major player. However, in recent years, the role of platelets has become controversial. Not only can this subcellular component release pro- and anti-inflammatory mediators, but it can also bind to different subsets of circulating lymphocytes, monocytes and neutrophils modulating their function in either direction. How platelets exert this dual role is not yet fully understood.
Collapse
Affiliation(s)
- Carlos Zamora
- Inflammatory Diseases, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Elisabet Cantó
- Inflammatory Diseases, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Sílvia Vidal
- Inflammatory Diseases, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| |
Collapse
|
14
|
Fu G, Deng M, Neal MD, Billiar TR, Scott MJ. Platelet-Monocyte Aggregates: Understanding Mechanisms and Functions in Sepsis. Shock 2021; 55:156-166. [PMID: 32694394 PMCID: PMC8008955 DOI: 10.1097/shk.0000000000001619] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
ABSTRACT Platelets have been shown to play an important immunomodulatory role in the pathogenesis of various diseases through their interactions with other immune and nonimmune cells. Sepsis is a major cause of death in the United States, and many of the mechanisms driving sepsis pathology are still unresolved. Monocytes have recently received increasing attention in sepsis pathogenesis, and multiple studies have associated increased levels of platelet-monocyte aggregates observed early in sepsis with clinical outcomes in sepsis patients. These findings suggest platelet-monocyte aggregates may be an important prognostic indicator. However, the mechanisms leading to platelet interaction and aggregation with monocytes, and the effects of aggregation during sepsis are still poorly defined. There are few studies that have really investigated functions of platelets and monocytes together, despite a large body of research showing separate functions of platelets and monocytes in inflammation and immune responses during sepsis. The goal of this review is to provide insights into what we do know about mechanisms and biological meanings of platelet-monocyte interactions, as well as some of the technical challenges and limitations involved in studying this important potential mechanism in sepsis pathogenesis. Improving our understanding of platelet and monocyte biology in sepsis may result in identification of novel targets that can be used to positively affect outcomes in sepsis.
Collapse
Affiliation(s)
- Guang Fu
- Department of General Surgery, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China (visiting scholar in Pittsburgh 2018-09/2020-09)
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Meihong Deng
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Matthew D. Neal
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Trauma Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Trauma Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Melanie J. Scott
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Trauma Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
15
|
Behl T, Sharma E, Sehgal A, Kaur I, Kumar A, Arora R, Pal G, Kakkar M, Kumar R, Bungau S. Expatiating the molecular approaches of HMGB1 in diabetes mellitus: Highlighting signalling pathways via RAGE and TLRs. Mol Biol Rep 2021; 48:1869-1881. [PMID: 33479829 DOI: 10.1007/s11033-020-06130-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/24/2020] [Indexed: 12/16/2022]
Abstract
Diabetes mellitus (DM) has become one of the major healthcare challenges worldwide in the recent times and inflammation being one of its key pathogenic process/mechanism affect several body parts including the peripheral and central nervous system. High-mobility group box 1 (HMGB1) is one of the major non-histone proteins that plays a key role in triggering the inflammatory response. Upon its release into the extracellular milieu, HMGB1 acts as an "alarmin" for the immune system to initiate tissue repair as a component of the host defense system. Furthermore, HMGB1 along with its downstream receptors like Toll-like receptors (TLRs) and receptors for advanced glycation end products (RAGE) serve as the suitable target for DM. The forthcoming research in the field of diabetes would potentially focus on the development of alternative approaches to target the centre of inflammation that is primarily mediated by HMGB1 to improve diabetic-related complications. This review covers the therapeutic actions of HMGB1 protein, which acts by activating the RAGE and TLR molecules to constitute a functional tripod system, in turn activating NF-κB pathway that contributes to the production of mediators for pro-inflammatory cytokines associated with DM. The interaction between TLR2 and TLR4 with ligands present in the host and the activation of RAGE stimulates various immune and metabolic responses that contribute to diabetes. This review emphasizes to elucidate the role of HMGB1 in the initiation and progression of DM and control over the inflammatory tripod as a promising therapeutic approach in the management of DM.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Eshita Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Rashmi Arora
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Giridhari Pal
- Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Munish Kakkar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ravinder Kumar
- Cardiovascular Research Institute, Icahn School of Medicine, New York, USA
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
16
|
Garman L, Pelikan RC, Rasmussen A, Lareau CA, Savoy KA, Deshmukh US, Bagavant H, Levin AM, Daouk S, Drake WP, Montgomery CG. Single Cell Transcriptomics Implicate Novel Monocyte and T Cell Immune Dysregulation in Sarcoidosis. Front Immunol 2020; 11:567342. [PMID: 33363531 PMCID: PMC7753017 DOI: 10.3389/fimmu.2020.567342] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/03/2020] [Indexed: 12/19/2022] Open
Abstract
Sarcoidosis is a systemic inflammatory disease characterized by infiltration of immune cells into granulomas. Previous gene expression studies using heterogeneous cell mixtures lack insight into cell-type-specific immune dysregulation. We performed the first single-cell RNA-sequencing study of sarcoidosis in peripheral immune cells in 48 patients and controls. Following unbiased clustering, differentially expressed genes were identified for 18 cell types and bioinformatically assessed for function and pathway enrichment. Our results reveal persistent activation of circulating classical monocytes with subsequent upregulation of trafficking molecules. Specifically, classical monocytes upregulated distinct markers of activation including adhesion molecules, pattern recognition receptors, and chemokine receptors, as well as enrichment of immunoregulatory pathways HMGB1, mTOR, and ephrin receptor signaling. Predictive modeling implicated TGFβ and mTOR signaling as drivers of persistent monocyte activation. Additionally, sarcoidosis T cell subsets displayed patterns of dysregulation. CD4 naïve T cells were enriched for markers of apoptosis and Th17/Treg differentiation, while effector T cells showed enrichment of anergy-related pathways. Differentially expressed genes in regulatory T cells suggested dysfunctional p53, cell death, and TNFR2 signaling. Using more sensitive technology and more precise units of measure, we identify cell-type specific, novel inflammatory and regulatory pathways. Based on our findings, we suggest a novel model involving four convergent arms of dysregulation: persistent hyperactivation of innate and adaptive immunity via classical monocytes and CD4 naïve T cells, regulatory T cell dysfunction, and effector T cell anergy. We further our understanding of the immunopathology of sarcoidosis and point to novel therapeutic targets.
Collapse
Affiliation(s)
- Lori Garman
- Oklahoma Medical Research Foundation, Genes and Human Disease, Oklahoma City, OK, United States
| | - Richard C Pelikan
- Oklahoma Medical Research Foundation, Genes and Human Disease, Oklahoma City, OK, United States
| | - Astrid Rasmussen
- Oklahoma Medical Research Foundation, Genes and Human Disease, Oklahoma City, OK, United States
| | - Caleb A Lareau
- Cell Circuits and Epigenomics Program, Broad Institute, Cambridge, MA, United States
| | - Kathryn A Savoy
- Oklahoma Medical Research Foundation, Genes and Human Disease, Oklahoma City, OK, United States
| | - Umesh S Deshmukh
- Oklahoma Medical Research Foundation, Arthritis and Clinical Immunology, Oklahoma City, OK, United States
| | - Harini Bagavant
- Oklahoma Medical Research Foundation, Arthritis and Clinical Immunology, Oklahoma City, OK, United States
| | - Albert M Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, United States
| | - Salim Daouk
- Cardiovascular Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Wonder P Drake
- Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Courtney G Montgomery
- Oklahoma Medical Research Foundation, Genes and Human Disease, Oklahoma City, OK, United States
| |
Collapse
|
17
|
Revisiting Platelets and Toll-Like Receptors (TLRs): At the Interface of Vascular Immunity and Thrombosis. Int J Mol Sci 2020; 21:ijms21176150. [PMID: 32858930 PMCID: PMC7504402 DOI: 10.3390/ijms21176150] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 12/19/2022] Open
Abstract
While platelet function has traditionally been described in the context of maintaining vascular integrity, recent evidence suggests that platelets can modulate inflammation in a much more sophisticated and nuanced manner than previously thought. Some aspects of this expanded repertoire of platelet function are mediated via expression of Toll-like receptors (TLRs). TLRs are a family of pattern recognition receptors that recognize pathogen-associated and damage-associated molecular patterns. Activation of these receptors is crucial for orchestrating and sustaining the inflammatory response to both types of danger signals. The TLR family consists of 10 known receptors, and there is at least some evidence that each of these are expressed on or within human platelets. This review presents the literature on TLR-mediated platelet activation for each of these receptors, and the existing understanding of platelet-TLR immune modulation. This review also highlights unresolved methodological issues that potentially contribute to some of the discrepancies within the literature, and we also suggest several recommendations to overcome these issues. Current understanding of TLR-mediated platelet responses in influenza, sepsis, transfusion-related injury and cardiovascular disease are discussed, and key outstanding research questions are highlighted. In summary, we provide a resource—a “researcher’s toolkit”—for undertaking further research in the field of platelet-TLR biology.
Collapse
|
18
|
Platelets Promote Macrophage Polarization toward Pro-inflammatory Phenotype and Increase Survival of Septic Mice. Cell Rep 2020; 28:896-908.e5. [PMID: 31340152 DOI: 10.1016/j.celrep.2019.06.062] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 02/28/2019] [Accepted: 06/17/2019] [Indexed: 12/21/2022] Open
Abstract
We investigated the contribution of human platelets to macrophage effector properties in the presence of lipopolysaccharide (LPS), as well as the beneficial effects and time frame for platelet transfusion in septic animals. Our results show that platelets sequester both pro-(TNF-α/IL-6) and anti-(IL-10) inflammatory cytokines released by monocytes. Low LPS concentrations (0.01 ng/mL) induced M2 macrophage polarization by decreasing CD64 and augmenting CD206 and CD163 expression; yet, the presence of platelets skewed monocytes toward type 1 macrophage (M1) phenotype in a cell-contact-dependent manner by the glycoprotein Ib (GPIb)-CD11b axis. Accordingly, platelet-licensed macrophages showed increased TNF-α levels, bacterial phagocytic activity, and a reduced healing capability. Platelet transfusion increased inducible nitric oxide synthase (iNOS)+ macrophages, improving bacterial clearance and survival rates in septic mice up to 6 h post-infection, an effect that was abolished by CD11b and GPIb blockade. Our results demonstrate that platelets orchestrate macrophage effector responses, improving the clinical outcome of sepsis in a narrow but relevant time frame.
Collapse
|
19
|
Zhai R, Blondonnet R, Ebrahimi E, Belville C, Audard J, Gross C, Choltus H, Henrioux F, Constantin JM, Pereira B, Blanchon L, Sapin V, Jabaudon M. The receptor for advanced glycation end-products enhances lung epithelial wound repair: An in vitro study. Exp Cell Res 2020; 391:112030. [PMID: 32330509 DOI: 10.1016/j.yexcr.2020.112030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/17/2020] [Accepted: 04/19/2020] [Indexed: 12/22/2022]
Abstract
Re-epithelialization of the alveolar surface is a key process of lung alveolar epithelial barrier repair after acute lung injury. The receptor for advanced glycation end-products (RAGE) pathway plays key roles in lung homeostasis, and its involvement in wound repair has been already reported in human bronchial epithelial cells. However, its effects on lung alveolar epithelial repair after injury remain unknown. We investigated whether RAGE stimulation with its ligands high-mobility group box 1 protein (HMGB1) or advanced glycation end-products (AGEs), alone or associated with RAGE inhibition using RAGE antagonist peptide, affects in vitro wound healing in human alveolar epithelial A549 cells. We further asked whether these effects could be associated with changes in cell proliferation and migration. We found that treatment of A549 cells with HMGB1 or AGEs promotes RAGE-dependent wound healing after a scratch assay. In addition, both RAGE ligands increased cell proliferation in a RAGE-dependent manner. Treatment with HMGB1 increased migration of alveolar epithelial cells at 12 h, independently of RAGE, whereas AGEs stimulated migration as measured 48 h after injury in a RAGE-dependent manner. Taken together, these results suggest that RAGE pathway is involved in lung alveolar epithelial wound repair, possibly through enhanced cell migration and proliferation.
Collapse
Affiliation(s)
- Ruoyang Zhai
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France
| | - Raiko Blondonnet
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France; Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Ebrahim Ebrahimi
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France; Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Corinne Belville
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France
| | - Jules Audard
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France; Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Christelle Gross
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France
| | - Helena Choltus
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France
| | - Fanny Henrioux
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France
| | - Jean-Michel Constantin
- Sorbonne University, GRC 29, AP-HP, DMU DREAM, Department of Anesthesiology and Critical Care, Pitié-Salpêtrière Hospital, Paris, France
| | - Bruno Pereira
- Biostatistics Unit, Department of Clinical Research and Innovation (DRCI), CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Loic Blanchon
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France
| | - Vincent Sapin
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France; Department of Medical Biochemistry and Molecular Genetics, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Matthieu Jabaudon
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France; Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France; Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
20
|
Egaña-Gorroño L, López-Díez R, Yepuri G, Ramirez LS, Reverdatto S, Gugger PF, Shekhtman A, Ramasamy R, Schmidt AM. Receptor for Advanced Glycation End Products (RAGE) and Mechanisms and Therapeutic Opportunities in Diabetes and Cardiovascular Disease: Insights From Human Subjects and Animal Models. Front Cardiovasc Med 2020; 7:37. [PMID: 32211423 PMCID: PMC7076074 DOI: 10.3389/fcvm.2020.00037] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 02/25/2020] [Indexed: 12/21/2022] Open
Abstract
Obesity and diabetes are leading causes of cardiovascular morbidity and mortality. Although extensive strides have been made in the treatments for non-diabetic atherosclerosis and its complications, for patients with diabetes, these therapies provide less benefit for protection from cardiovascular disease (CVD). These considerations spur the concept that diabetes-specific, disease-modifying therapies are essential to identify, especially as the epidemics of obesity and diabetes continue to expand. Hence, as hyperglycemia is a defining feature of diabetes, it is logical to probe the impact of the specific consequences of hyperglycemia on the vessel wall, immune cell perturbation, and endothelial dysfunction-all harbingers to the development of CVD. In this context, high levels of blood glucose stimulate the formation of the irreversible advanced glycation end products, the products of non-enzymatic glycation and oxidation of proteins and lipids. AGEs accumulate in diabetic circulation and tissues and the interaction of AGEs with their chief cellular receptor, receptor for AGE or RAGE, contributes to vascular and immune cell perturbation. The cytoplasmic domain of RAGE lacks endogenous kinase activity; the discovery that this intracellular domain of RAGE binds to the formin, DIAPH1, and that DIAPH1 is essential for RAGE ligand-mediated signal transduction, identifies the specific cellular means by which RAGE functions and highlights a new target for therapeutic interruption of RAGE signaling. In human subjects, prominent signals for RAGE activity include the presence and levels of two forms of soluble RAGE, sRAGE, and endogenous secretory (es) RAGE. Further, genetic studies have revealed single nucleotide polymorphisms (SNPs) of the AGER gene (AGER is the gene encoding RAGE) and DIAPH1, which display associations with CVD. This Review presents current knowledge regarding the roles for RAGE and DIAPH1 in the causes and consequences of diabetes, from obesity to CVD. Studies both from human subjects and animal models are presented to highlight the breadth of evidence linking RAGE and DIAPH1 to the cardiovascular consequences of these metabolic disorders.
Collapse
Affiliation(s)
- Lander Egaña-Gorroño
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Raquel López-Díez
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Gautham Yepuri
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Lisa S. Ramirez
- Department of Chemistry, University of Albany, State University of New York, Albany, NY, United States
| | - Sergey Reverdatto
- Department of Chemistry, University of Albany, State University of New York, Albany, NY, United States
| | - Paul F. Gugger
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Alexander Shekhtman
- Department of Chemistry, University of Albany, State University of New York, Albany, NY, United States
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
21
|
Le Y, Wang Y, Zhou L, Xiong J, Tian J, Yang X, Gai X, Sun Y. Cigarette smoke-induced HMGB1 translocation and release contribute to migration and NF-κB activation through inducing autophagy in lung macrophages. J Cell Mol Med 2020; 24:1319-1331. [PMID: 31769590 PMCID: PMC6991703 DOI: 10.1111/jcmm.14789] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 09/25/2019] [Accepted: 10/04/2019] [Indexed: 12/17/2022] Open
Abstract
High-mobility group box 1 (HMGB1) shows pro-inflammatory activity in various inflammatory diseases and has been found up-regulated in chronic obstructive pulmonary disease (COPD). Lung macrophages play an important role in airway inflammation and lung destruction in COPD, yet whether HMGB1 is involved in cigarette smoke (CS)-induced lung macrophage dysfunction is unknown. We sought to evaluate the intracellular localization and release of HMGB1 in lung macrophages from COPD patients and CS-exposed mice, and to investigate the role of HMGB1 in regulating autophagy in CS extract (CSE)-treated lung macrophages (MH-S cells). Our results showed that HMGB1 was highly expressed in lung tissues and sera of COPD patients and CS-exposed mice, along with predominantly cytoplasmic exporting from nuclei in lung macrophages. In vitro experiments revealed that CSE promoted the expression, nucleocytoplasmic translocation and release of HMGB1 partly via the nicotinic acetylcholine receptor (nAChR). Blockade of HMGB1 with chicken anti-HMGB1 polyclonal antibody (anti-HMGB1) or glycyrrhizin (Gly) attenuated the increase of LC3B-II and Beclin1, migration and p65 phosphorylation, suggesting the involvement of HMGB1 in autophagy, migration and NF-κB activation of lung macrophages. Hydroxychloroquine (CQ), an autophagy inhibitor, enhanced the increase of LC3B-II but not Beclin1 in CSE or rHMGB1-treated MH-S cells, and inhibition of autophagy by CQ and 3-methyladenine (3-MA) abrogated the migration and p65 phosphorylation of CSE-treated cells. These results indicate that CS-induced HMGB1 translocation and release contribute to migration and NF-κB activation through inducing autophagy in lung macrophages, providing novel evidence for HMGB1 as a potential target of intervention in COPD.
Collapse
Affiliation(s)
- Yanqing Le
- Department of Respiratory and Critical Care MedicinePeking University Third HospitalBeijingChina
| | - Yanhong Wang
- Department of Respiratory MedicineZhongshan City People's HospitalZhongshanChina
| | - Lu Zhou
- Department of Respiratory and Critical Care MedicinePeking University Third HospitalBeijingChina
| | - Jing Xiong
- Department of Respiratory and Critical Care MedicinePeking University Third HospitalBeijingChina
| | - Jieyu Tian
- Hematology Oncology CenterBeijing Children's HospitalCapital Medical UniversityBeijingChina
| | - Xia Yang
- Department of Respiratory MedicineTianjin Medical University General HospitalTianjingChina
| | - Xiaoyan Gai
- Department of Respiratory and Critical Care MedicinePeking University Third HospitalBeijingChina
| | - Yongchang Sun
- Department of Respiratory and Critical Care MedicinePeking University Third HospitalBeijingChina
| |
Collapse
|
22
|
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: 90] [Impact Index Per Article: 18.0] [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
|
23
|
de Carvalho GC, Borget MY, Bernier S, Garneau D, da Silva Duarte AJ, Dumais N. RAGE and CCR7 mediate the transmigration of Zika-infected monocytes through the blood-brain barrier. Immunobiology 2019; 224:792-803. [DOI: 10.1016/j.imbio.2019.08.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/12/2019] [Accepted: 08/22/2019] [Indexed: 12/13/2022]
|
24
|
Sun Z, Zhao M, Bian W, Ma H, Sun C. Associations of severity of fatty liver with oxidative stress, SAA, CRP and degree of cerebral arteriosclerosis in cerebral arteriosclerosis patients who have fatty liver. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:3022-3026. [PMID: 31934140 PMCID: PMC6949721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/22/2019] [Indexed: 06/10/2023]
Abstract
OBJECTIVE To analyze the associations of severity of fatty liver with oxidative stress, serum amyloid protein A (SAA), C-reactive protein (CRP) and degree of cerebral arteriosclerosis (CAS) in CAS patients with the complication of fatty liver, and to explore the predictive values of risk factors for these patients. METHODS A total of 200 patients diagnosed with CAS in our hospital from October 2016 to November 2018 were selected, including 90 cases with fatty liver (observation group) and 110 cases without fatty liver (control group), and there were 123 males and 77 females. The general clinical data, liver function, oxidative stress status, inflammatory factor levels, and degree of CAS were compared between the two groups, and their correlations and influencing factors were explored. RESULTS 1) There were no significant differences in the age, gender, and high-density lipoprotein (HDL) level between the observation group and control group (P>0.05). The body mass index (BMI), total cholesterol (TC), triglyceride (TG) and low-density lipoprotein (LDL) levels in the observation group were significantly higher than those in the control group. 2) The levels of liver function indexes, including alanine aminotransferase (ALT), aspartate aminotransferase (AST) and gamma-glutamyl transpeptidase (GGT), in the observation group were significantly higher than those in the control group, all (P<0.05). 3) In terms of the oxidative stress, the level of malondialdehyde (MDA) in the observation group was higher than that in the control group, while the levels of superoxide dismutase (SOD) and glutathione (GSH) in the observation group were obviously lower than in the control group, all (P<0.05). 4) The levels of inflammatory factors, including SAA, CRP and interleukin-6 (IL-6), in the observation group were much higher than those in the control group (P<0.05). 5) Observation group had higher whole blood low shear viscosity, whole blood high shear viscosity, and plasma viscosity than the control group (P<0.05). 6) ALT (r = 0.422, P = 0.000) and SAA (r = 0.828, P = 0.000) had positive correlations with the plasma viscosity, while GSH (r = -0.719, P<0.001) had a negative correlation with the plasma viscosity. CONCLUSION The liver function index levels, oxidative stress status, and inflammatory factor levels in CAS patients may affect the severity of arteriosclerosis and fatty liver.
Collapse
Affiliation(s)
- Zengqiang Sun
- Department of Neurology, Linzi District People’s HospitalZibo 255400, Shandong, China
| | - Mingzhi Zhao
- Department of Neurology, People’s Hospital of ZhongmuZhengzhou, Henan, China
| | - Wenwen Bian
- Department of Neurology, Linzi District People’s HospitalZibo 255400, Shandong, China
| | - Haifeng Ma
- Department of Emergency, Linzi District People’s HospitalZibo 255400, Shandong, China
| | - Chao Sun
- Department of Neurology, Linzi District People’s HospitalZibo 255400, Shandong, China
| |
Collapse
|
25
|
Goggs R, Jeffery U, LeVine DN, Li RHL. Neutrophil-Extracellular Traps, Cell-Free DNA, and Immunothrombosis in Companion Animals: A Review. Vet Pathol 2019; 57:6-23. [PMID: 31342866 DOI: 10.1177/0300985819861721] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Immunothrombosis is a potentially beneficial physiological process that aids innate immunity and host defense against pathogen invasion. However, this process can also be damaging when it occurs to excess or in critical blood vessels. Formation of extracellular traps by leukocytes, particularly neutrophils, is central to our understanding of immunothrombosis. In addition to degranulation and phagocytosis, extracellular traps are the third mechanism by which neutrophils combat potential pathogens. These traps consist of extracellular DNA decorated with bactericidal cellular proteins, including elastase, myeloperoxidase, and cathepsins. Neutrophils can release these structures as part of a controlled cell-death process or via a process termed vital NETosis that enables the cells to extrude DNA but remain viable. There is accumulating evidence that NETosis occurs in companion animals, including dogs, horses, and cats, and that it actively contributes to pathogenesis. Numerous studies have been published detailing various methods for identification and quantification of extracellular trap formation, including cell-free DNA, measurements of histones and proteins such as high-mobility group box-1, and techniques involving microscopy and flow cytometry. Here, we outline the present understanding of these phenomena and the mechanisms of extracellular trap formation. We critically review the data regarding measurement of NETosis in companion animals, summarize the existing literature on NETosis in veterinary species, and speculate on what therapeutic options these insights might present to clinicians in the future.
Collapse
Affiliation(s)
- Robert Goggs
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY, USA
| | - Unity Jeffery
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, USA
| | - Dana N LeVine
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| | - Ronald H L Li
- Department of Veterinary Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| |
Collapse
|
26
|
Gorgulho CM, Romagnoli GG, Bharthi R, Lotze MT. Johnny on the Spot-Chronic Inflammation Is Driven by HMGB1. Front Immunol 2019; 10:1561. [PMID: 31379812 PMCID: PMC6660267 DOI: 10.3389/fimmu.2019.01561] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 06/24/2019] [Indexed: 12/24/2022] Open
Abstract
Although much has been made of the role of HMGB1 acting as an acute damage associated molecular pattern (DAMP) molecule, prompting the response to tissue damage or injury, it is also released at sites of chronic inflammation including sites of infection, autoimmunity, and cancer. As such, the biology is distinguished from homeostasis and acute inflammation by the recruitment and persistence of myeloid derived suppressor cells, T regulatory cells, fibrosis and/or exuberant angiogenesis depending on the antecedents and the other individual inflammatory partners that HMGB1 binds and focuses, including IL-1β, CXCL12/SDF1, LPS, DNA, RNA, and sRAGE. High levels of HMGB1 released into the extracellular milieu and its persistence in the microenvironment can contribute to the pathogenesis of many if not all autoimmune disorders and is a key factor that drives inflammation further and worsens symptoms. HMGB1 is also pivotal in the maintenance of chronic inflammation and a “wound healing” type of immune response that ultimately contributes to the onset of carcinogenesis and tumor progression. Exosomes carrying HMGB1 and other instructive molecules are released and shape the response of various cells in the chronic inflammatory environment. Understanding the defining roles of REDOX, DAMPs and PAMPs, and the host response in chronic inflammation requires an alternative means for positing HMGB1's central role in limiting and focusing inflammation, distinguishing chronic from acute inflammation.
Collapse
Affiliation(s)
- Carolina M Gorgulho
- Tumor Immunology Laboratory, Department of Microbiology and Immunology, Botucatu Institute of Biosciences, São Paulo State University, Botucatu, Brazil.,DAMP Laboratory, Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Graziela G Romagnoli
- Tumor Immunology Laboratory, Department of Microbiology and Immunology, Botucatu Institute of Biosciences, São Paulo State University, Botucatu, Brazil
| | - Rosh Bharthi
- DAMP Laboratory, Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael T Lotze
- DAMP Laboratory, Department of Surgery, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Immunology, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
27
|
Ribeiro LS, Migliari Branco L, Franklin BS. Regulation of Innate Immune Responses by Platelets. Front Immunol 2019; 10:1320. [PMID: 31244858 PMCID: PMC6579861 DOI: 10.3389/fimmu.2019.01320] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/23/2019] [Indexed: 12/11/2022] Open
Abstract
The role of platelets has been extensively studied in the context of coagulation and vascular integrity. Their hemostatic imbalance can lead to known conditions as atherosclerotic plaques, thrombosis, and ischemia. Nevertheless, the knowledge regarding the regulation of different cell types by platelets has been growing exponentially in the past years. Among these biological systems, the innate immune response is remarkably affected by the crosstalk with platelets. This interaction can come from the formation of platelet-leukocyte aggregates, signaling by direct contact between membrane surface molecules or by the stimulation of immune cells by soluble factors and active microparticles secreted by platelets. These ubiquitous blood components are able to sense and react to danger signals, guiding leukocytes to an injury site and providing a scaffold for the formation of extracellular traps for efficient microbial killing and clearance. Using several different mechanisms, platelets have an important task as they regulate the release of different cytokines and chemokines upon sterile or infectious damage, the expression of cell markers and regulation of cell death and survival. Therefore, platelets are more than clotting agents, but critical players within the fine inflammatory equilibrium for the host. In this review, we present pointers to a better understanding about how platelets control and modulate innate immune cells, as well as a summary of the outcome of this interaction, providing an important step for therapeutic opportunities and guidance for future research on infectious and autoimmune diseases.
Collapse
Affiliation(s)
- Lucas Secchim Ribeiro
- Institute of Innate Immunity, University Hospitals, University of Bonn, Bonn, Germany
| | - Laura Migliari Branco
- Centro de Terapia Celular e Molecular (CTC-Mol), Universidade Federal de São Paulo, São Paulo, Brazil
| | - Bernardo S Franklin
- Institute of Innate Immunity, University Hospitals, University of Bonn, Bonn, Germany
| |
Collapse
|
28
|
Fujiwara M, Matoba T, Koga JI, Okahara A, Funamoto D, Nakano K, Tsutsui H, Egashira K. Nanoparticle incorporating Toll-like receptor 4 inhibitor attenuates myocardial ischaemia-reperfusion injury by inhibiting monocyte-mediated inflammation in mice. Cardiovasc Res 2019; 115:1244-1255. [PMID: 30851101 DOI: 10.1093/cvr/cvz066] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/22/2019] [Accepted: 03/07/2019] [Indexed: 01/03/2025] Open
Abstract
AIMS Myocardial ischaemia-reperfusion (IR) injury hampers the therapeutic effect of revascularization in patients with acute myocardial infarction (AMI). Innate immunity for damage-associated protein patterns promotes the process of IR injury; however, the blockade of Toll-like receptor 4 (TLR4) in myocardial IR injury has not been translated into clinical practice. Therefore, we aimed to examine whether the nanoparticle-mediated administration of TAK-242, a chemical inhibitor of TLR4, attenuates myocardial IR injury in a clinically feasible protocol in a mouse model. METHODS AND RESULTS We have prepared poly-(lactic-co-glycolic acid) nanoparticles containing TAK-242 (TAK-242-NP). TAK-242-NP significantly enhanced the drug delivery to monocytes/macrophages in the spleen, blood, and the heart in mice. Intravenous administration of TAK-242-NP (containing 1.0 or 3.0 mg/kg TAK-242) at the time of reperfusion decreased the infarct size, but the TAK-242 solution did not even when administered at a dosage of 10.0 mg/kg. TAK-242-NP inhibited the recruitment of Ly-6Chigh monocytes to the heart, which was accompanied by decreased circulating HMGB1, and NF-κB activation and cytokine expressions in the heart. TAK-242-NP did not decrease the infarct size further in TLR4-deficient mice, confirming the TLR4-specific mechanism in the effects of TAK-242-NP. Furthermore, TAK-242-NP did not decrease the infarct size further in CCR2-deficient mice, suggesting that monocyte/macrophage-mediated inflammation is the primary therapeutic target of TAK-242-NP. CONCLUSION The nanoparticle-mediated delivery of TAK-242-NP represent a novel and clinical feasible strategy in patients undergone coronary revascularization for AMI by regulating TLR4-dependent monocytes/macrophages-mediated inflammation.
Collapse
Affiliation(s)
- Masaki Fujiwara
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun-Ichiro Koga
- Department of Cardiovascular Research, Development, and Translational Research, Kyushu University, Fukuoka, Japan
| | - Arihide Okahara
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daiki Funamoto
- Department of Cardiovascular Research, Development, and Translational Research, Kyushu University, Fukuoka, Japan
| | - Kaku Nakano
- Department of Cardiovascular Research, Development, and Translational Research, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kensuke Egashira
- Department of Cardiovascular Research, Development, and Translational Research, Kyushu University, Fukuoka, Japan
| |
Collapse
|
29
|
Manganelli V, Truglia S, Capozzi A, Alessandri C, Riitano G, Spinelli FR, Ceccarelli F, Mancuso S, Garofalo T, Longo A, Valesini G, Sorice M, Conti F, Misasi R. Alarmin HMGB1 and Soluble RAGE as New Tools to Evaluate the Risk Stratification in Patients With the Antiphospholipid Syndrome. Front Immunol 2019; 10:460. [PMID: 30923525 PMCID: PMC6426766 DOI: 10.3389/fimmu.2019.00460] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/20/2019] [Indexed: 12/13/2022] Open
Abstract
Antiphospholipid antibody syndrome (APS) is a systemic autoimmune disease characterized by arterial and/or venous thrombosis, pregnancy morbidity in the presence of circulating “anti-phospholipid antibodies” (aPL). One of the main target antigens of aPL is β2-glycoprotein I (β2-GPI). APS may occur as a primary syndrome or associated with Systemic Lupus Erythematosus (SLE). High Mobility Group Box 1 (HMGB1) is a nuclear non-histone protein which is secreted from different type of cells during activation and/or cell death and may act as a proinflammatory mediator through ligation to its receptors, including RAGE. There is accumulating evidence that HMGB1 contributes to the pathogenesis of inflammatory and autoimmune diseases, especially SLE. In a previous study we demonstrated increased serum levels of HMGB1 in both primary and secondary APS patients. In this work we analyzed: (i) in vitro whether anti-β2-GPI antibodies from APS patients may induce both a HMGB1 cellular relocation by activation of its putative receptor RAGE in platelets and monocytes and, (ii) ex vivo, serum levels of HMGB1/soluble RAGE (sRAGE) in APS patients and their possible correlation with clinical manifestations. Platelets and monocytes from healthy donors were incubated with affinity purified anti-β2-GPI antibodies. HMGB1 and RAGE expression were analyzed by Western Blot. Sera from 60 consecutive APS patients (primary or secondary), diagnosed according to the Sydney Classification Criteria, were enrolled. As a control, 30 matched healthy subjects were studied. Serum levels of HMGB1 and sRAGE were analyzed by Western Blot. In vitro results showed that anti-β2-GPI antibodies were able to induce RAGE activation and HMGB1 cellular relocation in both monocytes and platelets. HMGB1 and sRAGE serum levels were significantly increased in APS patients in comparison with healthy subjects (p<0.0001). Interestingly, APS patients with spontaneous recurrent abortion showed significantly higher levels of sRAGE; moreover, in APS patients a direct correlation between serum levels of HMGB1 and disease duration was detected. Our observations suggest that anti-β2-GPI antibodies may trigger RAGE activation and HMGB1 cellular relocation during APS. Monitoring these molecules serum levels may represent an useful tool to evaluate the pathogenesis and risk stratification of clinical manifestations in APS.
Collapse
Affiliation(s)
- Valeria Manganelli
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| | - Simona Truglia
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Antonella Capozzi
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| | - Cristiano Alessandri
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Gloria Riitano
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| | - Francesca Romana Spinelli
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Fulvia Ceccarelli
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Silvia Mancuso
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Tina Garofalo
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| | - Agostina Longo
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| | - Guido Valesini
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Maurizio Sorice
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| | - Fabrizio Conti
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Roberta Misasi
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| |
Collapse
|
30
|
Inhibitory mechanisms of very low-dose rivaroxaban in non-ST-elevation myocardial infarction. Blood Adv 2019; 2:715-730. [PMID: 29588304 DOI: 10.1182/bloodadvances.2017013573] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 02/13/2018] [Indexed: 11/20/2022] Open
Abstract
Very low-dose (VLD) factor Xa (FXa) inhibition, in combination with acetylsalicylic acid (ASA) and clopidogrel, is associated with improved outcomes in patients with acute coronary syndrome (ACS) with a tolerable bleeding risk profile. To date, there are no data documenting platelet inhibition and the anticoagulatory effects of VLD FXa inhibition on top of guideline-adherent dual-antiplatelet therapy (DAPT) in patients with ACS. Patients with non-ST-elevation myocardial infarction (NSTEMI) receiving oral DAPT (ASA + clopidogrel, n = 20; or ASA + ticagrelor, n = 20) were prospectively enrolled in a nonrandomized study. Coagulation- and platelet-dependent thrombin generation (TG), measured by means of the calibrated automated thrombogram, were significantly decreased after in vitro and in vivo addition of rivaroxaban. As shown by a total thrombus-formation analysis approach, rivaroxaban treatment led to a significantly decreased coagulation-dependent (AR-chip) thrombus formation in patients treated with ASA plus P2Y12 inhibitor (clopidogrel/ticagrelor), whereas the pure platelet-dependent (PL-chip) thrombus formation was not affected at all. Adjunctive rivaroxaban therapy was not associated with significant differences in platelet aggregation assessed by light-transmission aggregometry (LTA). Nevertheless, according to fluorescence-activated cell sorter analysis, VLD rivaroxaban treatment resulted in a significantly reduced expression of platelet HMGB-1, whereas P-selectin exposure was not affected. Furthermore, an enhanced effect of rivaroxaban on total thrombus formation and TG was observed in particular in clopidogrel nonresponder patients defined as adenosine 5'-diphosphate-induced LTA ≥40%. VLD rivaroxaban reduces thrombus formation and platelet-dependent TG in patients with ACS receiving DAPT, which can be of potential ischemic benefit. This trial was registered at www.clinicaltrials.gov as #NCT01417884.
Collapse
|
31
|
|
32
|
Weng L, Guo L, Vachani A, Mesaros C, Blair IA. Quantification of Serum High Mobility Group Box 1 by Liquid Chromatography/High-Resolution Mass Spectrometry: Implications for Its Role in Immunity, Inflammation, and Cancer. Anal Chem 2018; 90:7552-7560. [PMID: 29791130 PMCID: PMC6417096 DOI: 10.1021/acs.analchem.8b01175] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
High mobility group
box 1 (HMGB1) is a non-histone chromosomal
protein, which can be secreted through a variety of pathways and bind
to pattern recognition receptors to release pro-inflammatory cytokines.
Previous studies have suggested that HMGB1 is upregulated in numerous
inflammatory diseases and that it could be a biomarker for such diseases.
However, these studies used immunoassay-based methods to analyze serum
HMGB1. Autoantibodies to HMGB1 in serum are found in healthy control
subjects as well as in patients with different diseases. HMGB1 also
binds to haptoglobin, a highly abundant plasma protein. This means
that antibodies used in immunoassays must compete with binding of
HMGB1 to endogenous serum HMGB1 autoantibodies and haptoglobin. To
overcome these potential problems, we developed and validated a specific
and sensitive assay based on stable isotope dilution and immunopurification
to quantify HMGB1 in plasma and serum using two-dimensional nano-ultra-high-performance
liquid chromatography parallel reaction monitoring/high-resolution
mass spectrometry. Using this assay, we found that serum HMGB1 in
24 healthy control subjects (6.0 ± 2.1 ng/mL) was above the mean
concentration reported for 18 different diseases (5.4 ± 2.8 ng/mL)
where the analyses were conducted with immunoassay methodology. In
light of our finding, the role of HMGB1 in these diseases will have
to be re-evaluated. The concentration of HMGB1 in citrated and EDTA-treated
plasma from the same healthy control subjects was below the limit
of detection of our assay (1 ng/mL), confirming that HMGB1 in serum
arises when blood is allowed to clot. This means that future studies
on the role of HMGB1 in vivo should be conducted on plasma rather
than serum.
Collapse
|
33
|
Yu SF, Feng WY, Chai SQ, Meng XB, Dou ZX, Zhu H. Down-Regulation of miR-218-5p Promotes Apoptosis of Human Umbilical Vein Endothelial Cells Through Regulating High-Mobility Group Box-1 in Henoch-Schonlein Purpura. Am J Med Sci 2018; 356:64-71. [PMID: 29751935 DOI: 10.1016/j.amjms.2018.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 04/02/2018] [Accepted: 04/04/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND Apoptosis of human umbilical vein endothelial cells (HUVECs) plays an important role in the progression of Henoch-Schonlein purpura (HSP). In the present study, we explored the function of miR-218-5p in HUVEC apoptosis and HSP development. MATERIALS AND METHODS HSP rat model was established and peripheral blood mononuclear cells (PBMC) were isolated. The expression of miR-218-5p and high-mobility group box-1 (HMGB1) protein in HUVECs was determined by quantitative real-time polymerase chain reaction and western blot, respectively. Cell apoptosis was detected by terminal deoxynucleotidyl transferase dUTP nick end labeling assay. The association between miR-218-5p and HMGB1 was determined by luciferase assay. The endogenous expression of related genes was modulated with recombinant plasmids and cell transfection. RESULTS MiR-218-5p was down-regulated and HMGB1 was up-regulated in vessels of the lower limb of HSP rats and in HUVECs co-cultured in HSP PBMC supernatant. MiR-218-5p negatively regulated HMGB1 by targeting its 3'-untranslated regions. Over expression of miR-218-5p reversed the increased apoptosis and HMGB1 expression observed in HUVECs co-cultured in PBMC supernatant, whereas miR-218-5p knockdown showed the opposite outcomes. Furthermore, the miR-218-5p mimic demonstrated an inhibitory effect on the apoptosis of HUVECs co-cultured in PBMC supernatant, which was reversed by over expression of HMGB1. In HSP rats, over expression of miR-218-5p attenuated HSP and decreased the level of HMGB1. CONCLUSIONS MiR-218-5p attenuated HSP at least partly through regulating HMGB1 expression and affecting the function of HUVECs.
Collapse
Affiliation(s)
- Shao-Fei Yu
- Department of Pediatrics, Inner Mongolia People's Hospital, Huhhot, Inner Mongolia, China.
| | - Wan-Yu Feng
- Department of Pediatrics, Inner Mongolia People's Hospital, Huhhot, Inner Mongolia, China
| | - Shao-Qing Chai
- Department of Pediatrics, Inner Mongolia People's Hospital, Huhhot, Inner Mongolia, China
| | - Xiao-Bo Meng
- Department of Pediatrics, Inner Mongolia People's Hospital, Huhhot, Inner Mongolia, China
| | - Zhong-Xia Dou
- Department of Pediatrics, Inner Mongolia People's Hospital, Huhhot, Inner Mongolia, China
| | - Hua Zhu
- Department of Pediatrics, Inner Mongolia People's Hospital, Huhhot, Inner Mongolia, China
| |
Collapse
|
34
|
Tan JY, Zhao F, Deng SX, Zhu HC, Gong Y, Wang W. Glycyrrhizin affects monocyte migration and apoptosis by blocking HMGB1 signaling. Mol Med Rep 2018; 17:5970-5975. [PMID: 29436639 DOI: 10.3892/mmr.2018.8598] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 01/29/2018] [Indexed: 11/05/2022] Open
Abstract
Monocytes serve an important role in systemic inflammation. High mobility group box‑1 protein (HMGB1) promotes recruitment and suppresses apoptosis in monocytes through the receptor for advanced glycation end products/ nuclear factor (NF)‑κB and toll‑like receptor 4/mitogen‑activated protein kinase (MAPK)/extracellular signal‑regulated kinase (ERK) signaling pathways. Glycyrrhizin (GL), an effective component of licorice, weakens the proinflammatory effect of HMGB1. The present study investigated the effect of GL on the migration and apoptosis of monocytes associated with HMGB1 signaling. THP‑1 cells were used to evaluate the behavior of monocytes in response to GL treatment, and the downstream pathways were investigated. GL suppressed HMGB1‑induced monocyte migration and increased HMGB1‑inhibited monocyte apoptosis. GL inhibited the activation of the NF‑κB and MAPK/ERK signaling pathways induced by HMGB1 and decreased the expression of monocyte chemoattractant protein‑1 (MCP‑1) and myeloid cell leukemia 1 (Mcl‑1). Taken together, the results indicated that GL may suppress the migration of monocytes and induce apoptosis to reduce systemic inflammation by blocking downstream NF‑κB/MCP‑1 and MAPK/ERK/Mcl‑1 signaling pathways.
Collapse
Affiliation(s)
- Jia-Ying Tan
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Feng Zhao
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Shui-Xiang Deng
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - He-Chen Zhu
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Ye Gong
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Wei Wang
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
35
|
Vogel S, Thein SL. Platelets at the crossroads of thrombosis, inflammation and haemolysis. Br J Haematol 2018; 180:761-767. [PMID: 29383704 DOI: 10.1111/bjh.15117] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Platelets play a critical role at the interphase of thrombosis and inflammation, key features in haemolysis-associated disorders. Exercising this role requires expression of pattern recognition receptors by platelets, including toll-like receptor 4 (TLR4) and nucleotide-binding domain leucine rich repeat containing protein 3 (NLRP3), the latter forming intraplatelet multiprotein inflammasome complexes. Platelets are a potential target of various damage-associated molecular pattern (DAMP) molecules, such as free haem, a degradation by-product of haemoglobin oxidation during haemolysis, and high-mobility group box 1 (HMGB1), a DNA-binding protein released by dying or stressed cells and activated platelets. We have recently identified platelet TLR4, NLRP3, and Bruton tyrosine kinase (BTK) as critical regulators of platelet aggregation and thrombus formation, suggesting that the BTK inhibitor ibrutinib is a potential therapeutic target. Increasing evidence suggests that these and other DAMP-driven signalling mechanisms employed by platelets might be key in mediating inflammation and thrombosis encountered in haemolytic disorders. However, the precise regulatory triggers and their clinical relevance are poorly understood. We provide new insights into these less-well characterised platelet mechanisms, which are potentially targetable in haemolytic disorders.
Collapse
Affiliation(s)
- Sebastian Vogel
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Swee Lay Thein
- Sickle Cell Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
36
|
The NLRP3 inflammasome and bruton's tyrosine kinase in platelets co-regulate platelet activation, aggregation, and in vitro thrombus formation. Biochem Biophys Res Commun 2016; 483:230-236. [PMID: 28034752 DOI: 10.1016/j.bbrc.2016.12.161] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 12/23/2016] [Indexed: 11/22/2022]
Abstract
Cleavage of interleukin-1β (IL-1β) is a key inflammatory event in immune cells and platelets, which is mediated by nucleotide-binding domain leucine rich repeat containing protein (NLRP3)-dependent activation of caspase-1. In immune cells, NLRP3 and caspase-1 form inflammasome complexes with the adaptor proteins apoptosis-associated speck-like protein containing a CARD (ASC) and bruton's tyrosine kinase (BTK). In platelets, however, the regulatory triggers and the functional effects of the NLRP3 inflammasome are unknown. Here, we show in vitro that the platelet NLRP3 inflammasome contributes to platelet activation, aggregation, and thrombus formation. NLRP3 activity, as monitored by caspase-1 activation and cleavage and secretion of IL-1β, was upregulated in activated platelets, which was dependent on platelet BTK. Pharmacological inhibition or genetic ablation of BTK in platelets led to decreased platelet activation, aggregation, and in vitro thrombus formation. We identify a functionally relevant link between BTK and NLRP3 in platelets, with potential implications in disease states associated with abnormal coagulation and inflammation.
Collapse
|