1
|
Hayashi H, Ishii M, Hasegawa Y, Taniguchi M. Critical pathomechanisms of NSAID-exacerbated respiratory disease (N-ERD) clarified by treatment with omalizumab, an anti-IgE antibody. Allergol Int 2025; 74:51-65. [PMID: 39419650 DOI: 10.1016/j.alit.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/18/2024] [Accepted: 08/23/2024] [Indexed: 10/19/2024] Open
Abstract
Characteristic symptoms of NSAID-exacerbated respiratory disease (N-ERD) include asthma, chronic eosinophilic rhinosinusitis with nasal polyposis, cysteinyl LT (CysLT) overproduction and NSAIDs hypersensitivity. Some N-ERD patients present with episodic treatment-resistant extra-respiratory symptoms (CysLT-associated coronary artery vasospasm, gastroenteritis, or skin rash). Even when using standard treatments for respiratory and extra-respiratory symptoms, including systemic corticosteroids and aspirin desensitization, it is difficult to control the clinical symptoms and severe type 2 inflammation involved with mast cells, eosinophils, ILC2s, and platelet activation. Few treatment options are applicable in a clinical setting. Therefore, identifying effective treatments is essential for managing N-ERD patients who suffer from these conditions. Our previous observational study demonstrated 12-month omalizumab treatment of N-ERD was clinically effective against respiratory symptoms. Despite the remaining eosinophilia, omalizumab significantly reduced urinary LTE4 and PGD2 metabolites to near normal levels at steady state. Based on the preliminary study, we demonstrated that omalizumab induced tolerance to aspirin in N-ERD patients 3 months after therapy initiation and suppressed activation of mast cells during 24 h of initiation in a randomized manner. Moreover, omalizumab had significant efficacy against extra-respiratory symptoms at baseline (lacking aspirin exposure) as well as throughout aspirin challenge. This review addresses the latest discoveries related to N-ERD pathogenesis and the significant effectiveness of omalizumab on N-ERD as a mast cell stabilizer. Our findings regarding omalizumab-associated mast cell inhibitory effects are indirect evidence that mast cell dysregulation and, possibly, IgE are pivotal components of N-ERD.
Collapse
Affiliation(s)
- Hiroaki Hayashi
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan; Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Makoto Ishii
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshinori Hasegawa
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan; National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Masami Taniguchi
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Sagamihara, Japan
| |
Collapse
|
2
|
Pan D, Ladds G, Rahman KM, Pitchford SC. Exploring bias in platelet P2Y 1 signalling: Host defence versus haemostasis. Br J Pharmacol 2024; 181:580-592. [PMID: 37442808 PMCID: PMC10952580 DOI: 10.1111/bph.16191] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/21/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Platelets are necessary for maintaining haemostasis. Separately, platelets are important for the propagation of inflammation during the host immune response against infection. The activation of platelets also causes inappropriate inflammation in various disease pathologies, often in the absence of changes to haemostasis. The separate functions of platelets during inflammation compared with haemostasis are therefore varied and this will be reflected in distinct pathways of activation. The activation of platelets by the nucleotide adenosine diphosphate (ADP) acting on P2Y1 and P2Y12 receptors is important for the development of platelet thrombi during haemostasis. However, P2Y1 stimulation of platelets is also important during the inflammatory response and paradoxically in scenarios where no changes to haemostasis and platelet aggregation occur. In these events, Rho-GTPase signalling, rather than the canonical phospholipase Cβ (PLCβ) signalling pathway, is necessary. We describe our current understanding of these differences, reflecting on recent advances in knowledge of P2Y1 structure, and the possibility of biased agonism occurring from activation via other endogenous nucleotides compared with ADP. Knowledge arising from these different pathways of P2Y1 stimulation of platelets during inflammation compared with haemostasis may help therapeutic control of platelet function during inflammation or infection, while preserving essential haemostasis. LINKED ARTICLES: This article is part of a themed issue on Platelet purinergic receptor and non-thrombotic disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.4/issuetoc.
Collapse
Affiliation(s)
- Dingxin Pan
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical ScienceKing's College LondonLondonUK
| | - Graham Ladds
- Department of PharmacologyUniversity of CambridgeCambridgeUK
| | - Khondaker Miraz Rahman
- Chemical Biology Group, Institute of Pharmaceutical ScienceKing's College LondonLondonUK
| | - Simon C. Pitchford
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical ScienceKing's College LondonLondonUK
| |
Collapse
|
3
|
Li D, Abhadiomhen SE, Zhou D, Shen XJ, Shi L, Cui Y. Asthma prediction via affinity graph enhanced classifier: a machine learning approach based on routine blood biomarkers. J Transl Med 2024; 22:100. [PMID: 38268004 PMCID: PMC10809685 DOI: 10.1186/s12967-024-04866-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 01/06/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Asthma is a chronic respiratory disease affecting millions of people worldwide, but early detection can be challenging due to the time-consuming nature of the traditional technique. Machine learning has shown great potential in the prompt prediction of asthma. However, because of the inherent complexity of asthma-related patterns, current models often fail to capture the correlation between data samples, limiting their accuracy. Our objective was to use our novel model to address the above problem via an Affinity Graph Enhanced Classifier (AGEC) to improve predictive accuracy. METHODS The clinical dataset used in this study consisted of 152 samples, where 24 routine blood markers were extracted as features to participate in the classification due to their ease of sourcing and relevance to asthma. Specifically, our model begins by constructing a projection matrix to reduce the dimensionality of the feature space while preserving the most discriminative features. Simultaneously, an affinity graph is learned through the resulting subspace to capture the internal relationship between samples better. Leveraging domain knowledge from the affinity graph, a new classifier (AGEC) is introduced for asthma prediction. AGEC's performance was compared with five state-of-the-art predictive models. RESULTS Experimental findings reveal the superior predictive capabilities of AGEC in asthma prediction. AGEC achieved an accuracy of 72.50%, surpassing FWAdaBoost (61.02%), MLFE (60.98%), SVR (64.01%), SVM (69.80%) and ERM (68.40%). These results provide evidence that capturing the correlation between samples can enhance the accuracy of asthma prediction. Moreover, the obtained [Formula: see text] values also suggest that the differences between our model and other models are statistically significant, and the effect of our model does not exist by chance. CONCLUSION As observed from the experimental results, advanced statistical machine learning approaches such as AGEC can enable accurate diagnosis of asthma. This finding holds promising implications for improving asthma management.
Collapse
Affiliation(s)
- Dejing Li
- Department of Respiratory, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
| | - Stanley Ebhohimhen Abhadiomhen
- School of Computer Science and Communication Engineering, JiangSu University, Zhenjiang, JiangSu, 212013, China
- Department of Computer Science, University of Nigeria, Nsukka, Nigeria
| | - Dongmei Zhou
- Clinical Research Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
| | - Xiang-Jun Shen
- School of Computer Science and Communication Engineering, JiangSu University, Zhenjiang, JiangSu, 212013, China
| | - Lei Shi
- Department of Clinical Laboratory, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, 201203, China.
| | - Yubao Cui
- Clinical Research Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China.
| |
Collapse
|
4
|
Foer D, Amin T, Nagai J, Tani Y, Feng C, Liu T, Newcomb DC, Lai J, Hayashi H, Snyder WE, McGill A, Lin A, Laidlaw T, Niswender KD, Boyce JA, Cahill KN. Glucagon-like Peptide-1 Receptor Pathway Attenuates Platelet Activation in Aspirin-Exacerbated Respiratory Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1806-1813. [PMID: 37870292 PMCID: PMC10842986 DOI: 10.4049/jimmunol.2300102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 10/03/2023] [Indexed: 10/24/2023]
Abstract
Platelets are key contributors to allergic asthma and aspirin-exacerbated respiratory disease (AERD), an asthma phenotype involving platelet activation and IL-33-dependent mast cell activation. Human platelets express the glucagon-like peptide-1 receptor (GLP-1R). GLP-1R agonists decrease lung IL-33 release and airway hyperresponsiveness in mouse asthma models. We hypothesized that GLP-1R agonists reduce platelet activation and downstream platelet-mediated airway inflammation in AERD. GLP-1R expression on murine platelets was assessed using flow cytometry. We tested the effect of the GLP-1R agonist liraglutide on lysine-aspirin (Lys-ASA)-induced changes in airway resistance, and platelet-derived mediator release in a murine AERD model. We conducted a prospective cohort study comparing the effect of pretreatment with liraglutide or vehicle on thromboxane receptor agonist-induced in vitro activation of platelets from patients with AERD and nonasthmatic controls. GLP-1R expression was higher on murine platelets than on leukocytes. A single dose of liraglutide inhibited Lys-ASA-induced increases in airway resistance and decreased markers of platelet activation and recruitment to the lung in AERD-like mice. Liraglutide attenuated thromboxane receptor agonist-induced activation as measured by CXCL7 release in plasma from patients with AERD and CD62P expression in platelets from both patients with AERD (n = 31) and nonasthmatic, healthy controls (n = 11). Liraglutide, a Food and Drug Administration-approved GLP-1R agonist for treatment of type 2 diabetes and obesity, attenuates in vivo platelet activation in an AERD murine model and in vitro activation in human platelets in patients with and without AERD. These data advance the GLP-1R axis as a new target for platelet-mediated inflammation warranting further study in asthma.
Collapse
Affiliation(s)
- Dinah Foer
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA
| | - Taneem Amin
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jun Nagai
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA
| | - Yumi Tani
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Chunli Feng
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA
| | - Tao Liu
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA
| | - Dawn C. Newcomb
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - Juying Lai
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Hiroaki Hayashi
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - William E. Snyder
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alanna McGill
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Anabel Lin
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tanya Laidlaw
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA
| | - Kevin D. Niswender
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- United States Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Joshua A. Boyce
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA
| | - Katherine N. Cahill
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
5
|
Gruba S, Wu X, Spanolios E, He J, Xiong-Hang K, Haynes CL. Platelet Response to Allergens, CXCL10, and CXCL5 in the Context of Asthma. ACS BIO & MED CHEM AU 2023; 3:87-96. [PMID: 36820311 PMCID: PMC9936497 DOI: 10.1021/acsbiomedchemau.2c00059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022]
Abstract
Asthma is a chronic respiratory disease initiated by a variety of factors, including allergens. During an asthma attack, the secretion of C-X-C-motif chemokine 10 (CXCL10) and chemokine ligand 5 (CCL5) causes the migration of immune cells, including platelets, into the lungs and airway. Platelets, which contain three classes of chemical messenger-filled granules, can secrete vasodilators (adenosine diphosphate and adenosine triphosphate), serotonin (a vasoconstrictor and a vasodilator, depending on the biological system), platelet-activating factor, N-formylmethionyl-leucyl-phenylalanine ((fMLP), a bacterial tripeptide that stimulates chemotaxis), and chemokines (CCL5, platelet factor 4 (PF4), and C-X-C-motif chemokine 12 (CXCL12)), amplifying the asthma response. The goal of this work was threefold: (1) to understand if and how the antibody immunoglobulin E (IgE), responsible for allergic reactions, affects platelet response to the common platelet activator thrombin; (2) to understand how allergen stimulation compares to thrombin stimulation; and (3) to monitor platelet response to fMLP and the chemokines CXCL10 and CCL5. Herein, high-pressure liquid chromatography with electrochemical detection and/or carbon-fiber microelectrode amperometry measured granular secretion events from platelets with and without IgE in the presence of the allergen 2,4,6-trinitrophenyl-conjugated ovalbumin (TNP-Ova), thrombin, CXCL10, or CCL5. Platelet adhesion and chemotaxis were measured using a microfluidic platform in the presence of CXCL10, CCL5, or TNP-OVA. Results indicate that IgE binding promotes δ-granule secretion in response to platelet stimulation by thrombin in bulk. Single-cell results on platelets with exogenous IgE exposure showed significant changes in the post-membrane-granule fusion behavior during chemical messenger delivery events after thrombin stimulation. In addition, TNP-Ova allergen stimulation of IgE-exposed platelets secreted serotonin to the same extent as thrombin platelet stimulation. Enhanced adhesion to endothelial cells was demonstrated by TNP-Ova stimulation. Finally, only after incubation with IgE did platelets secrete chemical messengers in response to stimulation with fMLP, CXCL10, and CCL5.
Collapse
Affiliation(s)
- Sarah Gruba
- Department of Chemistry, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Xiaojie Wu
- Department of Chemistry, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Eleni Spanolios
- Department of Chemistry, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jiayi He
- Department of Chemistry, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Kang Xiong-Hang
- Department of Chemistry, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Christy L. Haynes
- Department of Chemistry, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
6
|
Abstract
In addition to the key role in hemostasis and thrombosis, platelets have also been wildly acknowledged as immune regulatory cells and involving in the pathogenesis of inflammation-related diseases. Since purine receptor P2Y12 plays a crucial role in platelet activation, P2Y12 antagonists such as clopidogrel, prasugrel, and ticagrelor have been widely used in cardiovascular diseases worldwide in recent decades due to their potent antiplatelet and antithrombotic effects. Meanwhile, the role of P2Y12 in inflammatory diseases has also been extensively studied. Relatively, there are few studies on the regulation of P2Y12. This review first summarizes the various roles of P2Y12 in the process of platelet activation, as well as downstream effects and signaling pathways; then introduces the effects of P2Y12 in inflammatory diseases such as sepsis, atherosclerosis, cancer, autoimmune diseases, and asthma; and finally reviews the current researches on P2Y12 regulation.
Collapse
Affiliation(s)
- Xiaohua Li
- Department of Infectious Diseases, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
- Department of Pharmacology, School of Pharmacy, Jilin University, Fujin Road, Changchun, 130021, Jilin, China
| | | | - Xia Cao
- Department of Pharmacology, School of Pharmacy, Jilin University, Fujin Road, Changchun, 130021, Jilin, China.
| |
Collapse
|
7
|
Varga I, Michalka P, Mištinová JP. Complications after administration of mRNA vaccine against COVID-19 - case report and short review. VNITRNI LEKARSTVI 2023; 69:20-27. [PMID: 37468319 DOI: 10.36290/vnl.2023.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
The pandemic of the disease COVID-19 (COronaVIrus Disease 2019) caused by the SARS-CoV-2 coronavirus (severe acute respiratory syndrome coronavirus 2) resulted in millions of deaths and many patients have chronic consequences after overcoming the acute condition. Several vaccines have been developed in an effort to stop the spread of the virus, but they have potentially serious adverse effects. We present a case report of a patient with acute (myocarditis, exacerbation of bronchial asthma) and long-term (postural orthostatic tachycardia syndrome - POTS) complications after vaccination with the second dose of mRNA vaccine BNT162b2 (Comirnaty®). Treatment consists of regimen measures, numerous pharmacotherapy (metoprolol, ivabradine, corticosteroids, antihistamines, antiphlogistics, bronchodilators) and several nutraceuticals (maritime pine bark extract, quercetin, vitamins, magnesium, phosphatidylcholine). In the discussion, we analyze post-vaccination injury and present a short review of the current literature.
Collapse
|
8
|
McGrath FM, Francis A, Fatovich DM, Macdonald SPJ, Arendts G, Woo AJ, Bosio E. Genes involved in platelet aggregation and activation are downregulated during acute anaphylaxis in humans. Clin Transl Immunology 2022; 11:e1435. [PMID: 36583159 PMCID: PMC9791329 DOI: 10.1002/cti2.1435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/20/2022] [Accepted: 12/05/2022] [Indexed: 12/27/2022] Open
Abstract
Objective Mechanisms underlying the anaphylactic reaction in humans are not fully understood. Here, we aimed at improving our understanding of anaphylaxis by investigating gene expression changes. Methods Microarray data set GSE69063 was analysed, describing emergency department (ED) patients with severe anaphylaxis (n = 12), moderate anaphylaxis (n = 6), sepsis (n = 20) and trauma (n = 11). Samples were taken at ED presentation (T0) and 1 h later (T1). Healthy controls were age and sex matched to ED patient groups. Gene expression changes were determined using limma, and pathway analysis applied. Differentially expressed genes were validated in an independent cohort of anaphylaxis patients (n = 31) and matched healthy controls (n = 10), using quantitative reverse transcription-polymerase chain reaction. Results Platelet aggregation was dysregulated in severe anaphylaxis at T0, but not in moderate anaphylaxis, sepsis or trauma. Dysregulation was not observed in patients who received adrenaline before T0. Seven genes (GATA1 (adjusted P-value = 5.57 × 10-4), TLN1 (adjusted P-value = 9.40 × 10-4), GP1BA (adjusted P-value = 2.15 × 10-2), SELP (adjusted P-value = 2.29 × 10-2), MPL (adjusted P-value = 1.20 × 10-2), F13A1 (adjusted P-value = 1.39 × 10-2) and SPARC (adjusted P-value = 4.06 × 10-2)) were significantly downregulated in severe anaphylaxis patients who did not receive adrenaline before ED arrival, compared with healthy controls. One gene (TLN1 (adjusted P-value = 1.29 × 10-2)) was significantly downregulated in moderate anaphylaxis patients who did not receive adrenaline before ED arrival, compared with healthy controls. Conclusion Downregulation of genes involved in platelet aggregation and activation is a unique feature of the early anaphylactic reaction not previously reported and may be associated with reaction severity.
Collapse
Affiliation(s)
- Francesca M McGrath
- Centre for Clinical Research in Emergency MedicineHarry Perkins Institute of Medical ResearchPerthWAAustralia
| | - Abbie Francis
- Centre for Clinical Research in Emergency MedicineHarry Perkins Institute of Medical ResearchPerthWAAustralia,Telethon Kids Institute, Centre for Child Health Research, The University of Western AustraliaNedlandsWAAustralia
| | - Daniel M Fatovich
- Centre for Clinical Research in Emergency MedicineHarry Perkins Institute of Medical ResearchPerthWAAustralia,Discipline of Emergency Medicine, Medical SchoolUniversity of Western AustraliaPerthWAAustralia,Emergency DepartmentRoyal Perth HospitalPerthWAAustralia
| | - Stephen PJ Macdonald
- Centre for Clinical Research in Emergency MedicineHarry Perkins Institute of Medical ResearchPerthWAAustralia,Discipline of Emergency Medicine, Medical SchoolUniversity of Western AustraliaPerthWAAustralia,Emergency DepartmentRoyal Perth HospitalPerthWAAustralia
| | - Glenn Arendts
- Centre for Clinical Research in Emergency MedicineHarry Perkins Institute of Medical ResearchPerthWAAustralia,Discipline of Emergency Medicine, Medical SchoolUniversity of Western AustraliaPerthWAAustralia,Emergency DepartmentFiona Stanley HospitalPerthWAAustralia
| | - Andrew J Woo
- Laboratory for Cancer MedicineHarry Perkins Institute of Medical ResearchPerthWAAustralia,School of Medical and Health SciencesEdith Cowan UniversityPerthWAAustralia
| | - Erika Bosio
- Centre for Clinical Research in Emergency MedicineHarry Perkins Institute of Medical ResearchPerthWAAustralia,Discipline of Emergency Medicine, Medical SchoolUniversity of Western AustraliaPerthWAAustralia
| |
Collapse
|
9
|
Gu C, Loube J, Lee R, Bevans-Fonti S, Wu TD, Barmine JH, Jun JC, McCormack MC, Hansel NN, Mitzner W, Polotsky VY. Metformin Alleviates Airway Hyperresponsiveness in a Mouse Model of Diet-Induced Obesity. Front Physiol 2022; 13:883275. [PMID: 35574481 PMCID: PMC9098833 DOI: 10.3389/fphys.2022.883275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/14/2022] [Indexed: 12/03/2022] Open
Abstract
Obese asthma is a unique phenotype of asthma characterized by non-allergic airway hyperresponsiveness (AHR) and inflammation which responds poorly to standard asthma therapy. Metformin is an oral hypoglycemic drug with insulin-sensitizing and anti-inflammatory properties. The objective of the current study was to test the effect of metformin on AHR in a mouse model of diet-induced obesity (DIO). We fed 12-week-old C57BL/6J DIO mice with a high fat diet for 8 weeks and treated them with either placebo (control, n = 10) or metformin (n = 10) added in drinking water (300 mg/kg/day) during the last 2 weeks of the experiment. We assessed AHR, metabolic profiles, and inflammatory markers after treatments. Metformin did not affect body weight or fasting blood glucose, but significantly reduced serum insulin (p = 0.0117). Metformin reduced AHR at 30 mg/ml of methacholine challenge (p = 0.0052) without affecting baseline airway resistance. Metformin did not affect circulating white blood cell counts or lung cytokine mRNA expression, but modestly decreased circulating platelet count. We conclude that metformin alleviated AHR in DIO mice. This finding suggests metformin has the potential to become an adjuvant pharmacological therapy in obese asthma.
Collapse
Affiliation(s)
- Chenjuan Gu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jeff Loube
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Rachel Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shannon Bevans-Fonti
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Tianshi David Wu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine and the Center for Innovations in Quality, Effectiveness, and Safety, Michael E. DeBakey VA Medical Center, Houston, TX, United States
| | - Jessica H. Barmine
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jonathan C. Jun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Meredith C. McCormack
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nadia N. Hansel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Wayne Mitzner
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Vsevolod Y. Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- *Correspondence: Vsevolod Y. Polotsky,
| |
Collapse
|
10
|
Orimo K, Tamari M, Takeda T, Kubo T, Rückert B, Motomura K, Sugiyama H, Yamada A, Saito K, Arae K, Kuriyama M, Hara M, Soyka MB, Ikutani M, Yamaguchi S, Morimoto N, Nakabayashi K, Hata K, Matsuda A, Akdis CA, Sudo K, Saito H, Nakae S, Tamaoki J, Tagaya E, Matsumoto K, Morita H. Direct platelet adhesion potentiates group 2 innate lymphoid cell functions. Allergy 2022; 77:843-855. [PMID: 34402091 DOI: 10.1111/all.15057] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 08/11/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Platelets are thought to be involved in the pathophysiology of asthma, presumably through direct adhesion to inflammatory cells, including group 2 innate lymphoid cells (ILC2s). Here, we tried to elucidate the effects of platelet adhesion to ILC2s in vitro and in vivo, as well as the mechanisms involved. METHODS Alternaria-induced ILC2-dependent airway inflammation models using wild-type and c-mpl-/- mice were evaluated. Both purified CD41+ and CD41- ILC2s were cultured with IL-2 and IL-33 to determine in vitro Type 2 (T2) cytokine production and cell proliferation. RNA-seq data of flow-cytometry-sorted CD41+ and CD41- ILC2s were used to isolate ILC2-specific genes. Flow cytometry was performed to determine the expression of CD41 and adhesion-related molecules on ILC2s in both mouse and human tissues. RESULTS T2 inflammation and T2 cytokine production from ILC2s were significantly reduced in the c-mpl-/- mice compared to wild-type mice. Platelet-adherent ILC2s underwent significant proliferation and showed enhanced T2 cytokine production when exposed to IL-2 and IL-33. The functions of ILC2-specific genes were related to cell development and function. Upstream regulator analysis identified 15 molecules, that are thought to be involved in ILC2 activation. CD41 expression levels were higher in ILC2s from human PBMCs and mouse lung than in those from secondary lymphoid tissues, but they did not correlate with the P-selectin glycoprotein ligand-1 or CD24 expression level. CONCLUSION Platelets spontaneously adhere to ILC2s, probably in the peripheral blood and airways, thereby potentiating ILC2s to enhance their responses to IL-33.
Collapse
Affiliation(s)
- Keisuke Orimo
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
- Department of Respiratory Medicine Tokyo Women's Medical University Tokyo Japan
| | - Masato Tamari
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
- Department of Pediatrics Jikei University School of Medicine Tokyo Japan
| | - Tomohiro Takeda
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
- Department of Health Science Kansai University of Health Sciences Osaka Japan
| | - Terufumi Kubo
- Department of Pathology Sapporo Medical University School of Medicine Sapporo Japan
| | - Beate Rückert
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Kenichiro Motomura
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Hiroki Sugiyama
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Ayako Yamada
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Kyoko Saito
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
- Department of Otorhinolaryngology Head and Neck Surgery University of Fukui Fukui Japan
| | - Ken Arae
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
- Department of Immunology Faculty of Health Sciences Kyorin University Tokyo Japan
| | - Motohiro Kuriyama
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Mariko Hara
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Michael B. Soyka
- Department of Otorhinolaryngology, Head and Neck Surgery University Hospital Zurich and University of Zurich Zurich Switzerland
| | - Masashi Ikutani
- Graduate School of Integrated Sciences for Life Hiroshima University Higashi‐Hiroshima City Japan
- Department of Immune Regulation Research Institute, National Center for Global Health and Medicine Ichikawa Japan
| | - Sota Yamaguchi
- Division of Otolaryngology Department of Surgical Specialties National Center for Child Health and Development Tokyo Japan
| | - Noriko Morimoto
- Division of Otolaryngology Department of Surgical Specialties National Center for Child Health and Development Tokyo Japan
| | - Kazuhiko Nakabayashi
- Department of Maternal–Fetal Biology National Research Institute for Child Health and Development Tokyo Japan
| | - Kenichiro Hata
- Department of Maternal–Fetal Biology National Research Institute for Child Health and Development Tokyo Japan
| | - Akio Matsuda
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Katsuko Sudo
- Animal Research Center Tokyo Medical University Tokyo Japan
| | - Hirohisa Saito
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Susumu Nakae
- Department of Immune Regulation Research Institute, National Center for Global Health and Medicine Ichikawa Japan
- Laboratory of Systems Biology Center for Experimental Medicine and Systems Biology The Institute of Medical Science, The University of Tokyo Tokyo Japan
- Precursory Research for Embryonic Science and Technology (PRESTO Japan Science and Technology Agency Saitama Japan
| | - Jun Tamaoki
- Department of Respiratory Medicine Tokyo Women's Medical University Tokyo Japan
| | - Etsuko Tagaya
- Department of Respiratory Medicine Tokyo Women's Medical University Tokyo Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Hideaki Morita
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
- Allergy Center National Center for Child Health and Development Tokyo Japan
| |
Collapse
|
11
|
Tan XY, Jing HY, Ma YR. Interleukin-33/ Suppression of Tumorigenicity 2 in Renal Fibrosis: Emerging Roles in Prognosis and Treatment. Front Physiol 2022; 12:792897. [PMID: 35046838 PMCID: PMC8761767 DOI: 10.3389/fphys.2021.792897] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/02/2021] [Indexed: 12/20/2022] Open
Abstract
Chronic kidney disease (CKD) is a major public health problem that affects more than 10% of the population worldwide and has a high mortality rate. Therefore, it is necessary to identify novel treatment strategies for CKD. Incidentally, renal fibrosis plays a central role in the progression of CKD to end-stage renal disease (ESRD). The activation of inflammatory pathways leads to the development of renal fibrosis. In fact, interleukin-33 (IL-33), a newly discovered member of the interleukin 1 (IL-1) cytokine family, is a crucial regulator of the inflammatory process. It exerts pro-inflammatory and pro-fibrotic effects via the suppression of tumorigenicity 2 (ST2) receptor, which, in turn, activates other inflammatory pathways. Although the role of this pathway in cardiac, pulmonary, and hepatic fibrotic diseases has been extensively studied, its precise role in renal fibrosis has not yet been completely elucidated. Recent studies have shown that a sustained activation of IL-33/ST2 pathway promotes the development of renal fibrosis. However, with prolonged research in this field, it is expected that the IL-33/ST2 pathway will be used as a diagnostic and prognostic tool for renal diseases. In addition, the IL-33/ST2 pathway seems to be a new target for the future treatment of CKD. Here, we review the mechanisms and potential applications of the IL-33/ST2 pathway in renal fibrosis; such that it can help clinicians and researchers to explore effective treatment options and develop novel medicines for CKD patients.
Collapse
Affiliation(s)
- Xiao-Yang Tan
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hao-Yue Jing
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue-Rong Ma
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
12
|
Atypical Roles of the Chemokine Receptor ACKR3/CXCR7 in Platelet Pathophysiology. Cells 2022; 11:cells11020213. [PMID: 35053329 PMCID: PMC8773869 DOI: 10.3390/cells11020213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/23/2022] Open
Abstract
The manifold actions of the pro-inflammatory and regenerative chemokine CXCL12/SDF-1α are executed through the canonical GProteinCoupledReceptor CXCR4, and the non-canonical ACKR3/CXCR7. Platelets express CXCR4, ACKR3/CXCR7, and are a vital source of CXCL12/SDF-1α themselves. In recent years, a regulatory impact of the CXCL12-CXCR4-CXCR7 axis on platelet biogenesis, i.e., megakaryopoiesis, thrombotic and thrombo-inflammatory actions have been revealed through experimental and clinical studies. Platelet surface expression of ACKR3/CXCR7 is significantly enhanced following myocardial infarction (MI) in acute coronary syndrome (ACS) patients, and is also associated with improved functional recovery and prognosis. The therapeutic implications of ACKR3/CXCR7 in myocardial regeneration and improved recovery following an ischemic episode, are well documented. Cardiomyocytes, cardiac-fibroblasts, endothelial lining of the blood vessels perfusing the heart, besides infiltrating platelets and monocytes, all express ACKR3/CXCR7. This review recapitulates ligand induced differential trafficking of platelet CXCR4-ACKR3/CXCR7 affecting their surface availability, and in regulating thrombo-inflammatory platelet functions and survival through CXCR4 or ACKR3/CXCR7. It emphasizes the pro-thrombotic influence of CXCL12/SDF-1α exerted through CXCR4, as opposed to the anti-thrombotic impact of ACKR3/CXCR7. Offering an innovative translational perspective, this review also discusses the advantages and challenges of utilizing ACKR3/CXCR7 as a potential anti-thrombotic strategy in platelet-associated cardiovascular disorders, particularly in coronary artery disease (CAD) patients post-MI.
Collapse
|
13
|
Jahan P, Tahseen R, Parvez M, Kumar GS. A correlational study on neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio in bronchial asthma. ADVANCES IN HUMAN BIOLOGY 2022. [DOI: 10.4103/aihb.aihb_44_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
14
|
Chebbo M, Duez C, Alessi MC, Chanez P, Gras D. Platelets: a potential role in chronic respiratory diseases? Eur Respir Rev 2021; 30:30/161/210062. [PMID: 34526315 PMCID: PMC9488457 DOI: 10.1183/16000617.0062-2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/05/2021] [Indexed: 12/21/2022] Open
Abstract
Platelets are small anucleate cells known for their role in haemostasis and thrombosis. In recent years, an increasing number of observations have suggested that platelets are also immune cells and key modulators of immunity. They express different receptors and molecules that allow them to respond to pathogens, and to interact with other immune cells. Platelets were linked to the pathogenesis of some inflammatory disorders including respiratory diseases such as asthma and idiopathic pulmonary fibrosis. Here, we discuss the involvement of platelets in different immune responses, and we focus on their potential role in various chronic lung diseases. In addition to their essential role in haemostasis and thrombosis, platelets are strong modulators of different immune responses, and could be involved in the physiopathology of several chronic airway diseaseshttps://bit.ly/3cB6Xnj
Collapse
Affiliation(s)
| | | | - Marie C Alessi
- Aix-Marseille Univ, INSERM, INRAE, Marseille, France.,APHM, CHU de la Timone, Laboratoire d'hématologie, Marseille, France
| | - Pascal Chanez
- Aix-Marseille Univ, INSERM, INRAE, Marseille, France.,APHM, Hôpital NORD, Clinique des Bronches, Allergie et Sommeil, Marseille, France
| | - Delphine Gras
- Aix-Marseille Univ, INSERM, INRAE, Marseille, France
| |
Collapse
|
15
|
Banić I, Lovrić M, Cuder G, Kern R, Rijavec M, Korošec P, Turkalj M. Treatment outcome clustering patterns correspond to discrete asthma phenotypes in children. Asthma Res Pract 2021; 7:11. [PMID: 34344475 PMCID: PMC8330019 DOI: 10.1186/s40733-021-00077-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/18/2021] [Indexed: 11/10/2022] Open
Abstract
Despite widely and regularly used therapy asthma in children is not fully controlled. Recognizing the complexity of asthma phenotypes and endotypes imposed the concept of precision medicine in asthma treatment. By applying machine learning algorithms assessed with respect to their accuracy in predicting treatment outcome, we have successfully identified 4 distinct clusters in a pediatric asthma cohort with specific treatment outcome patterns according to changes in lung function (FEV1 and MEF50), airway inflammation (FENO) and disease control likely affected by discrete phenotypes at initial disease presentation, differing in the type and level of inflammation, age of onset, comorbidities, certain genetic and other physiologic traits. The smallest and the largest of the 4 clusters- 1 (N = 58) and 3 (N = 138) had better treatment outcomes compared to clusters 2 and 4 and were characterized by more prominent atopic markers and a predominant allelic (A allele) effect for rs37973 in the GLCCI1 gene previously associated with positive treatment outcomes in asthmatics. These patients also had a relatively later onset of disease (6 + yrs). Clusters 2 (N = 87) and 4 (N = 64) had poorer treatment success, but varied in the type of inflammation (predominantly neutrophilic for cluster 4 and likely mixed-type for cluster 2), comorbidities (obesity for cluster 2), level of systemic inflammation (highest hsCRP for cluster 2) and platelet count (lowest for cluster 4). The results of this study emphasize the issues in asthma management due to the overgeneralized approach to the disease, not taking into account specific disease phenotypes.
Collapse
Affiliation(s)
- Ivana Banić
- Srebrnjak Children's Hospital, Srebrnjak 100, 10000, Zagreb, Croatia
| | - Mario Lovrić
- Know-Center, Infeldgasse 13, Graz, AT-8010, Austria. .,Institute of Interactive Systems and Data Science, Graz University of Technology, Inffeldgasse 16C, AT-8010, Graz, Austria.
| | - Gerald Cuder
- Institute of Interactive Systems and Data Science, Graz University of Technology, Inffeldgasse 16C, AT-8010, Graz, Austria
| | - Roman Kern
- Know-Center, Infeldgasse 13, Graz, AT-8010, Austria.,Institute of Interactive Systems and Data Science, Graz University of Technology, Inffeldgasse 16C, AT-8010, Graz, Austria
| | - Matija Rijavec
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik 36, 4204, Golnik, Slovenia
| | - Peter Korošec
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik 36, 4204, Golnik, Slovenia
| | - Mirjana Turkalj
- Srebrnjak Children's Hospital, Srebrnjak 100, 10000, Zagreb, Croatia.,Faculty of Medicine, J.J, Strossmayer University of Osijek, Josipa Huttlera 4, 31000, Osijek, Croatia.,Catholic University of Croatia, Ilica 242, 10000, Zagreb, Croatia
| |
Collapse
|
16
|
Amison RT, Page CP. Novel pharmacological therapies for the treatment of bronchial asthma. Minerva Med 2021; 113:31-50. [PMID: 34236157 DOI: 10.23736/s0026-4806.21.07559-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Asthma has long been recognised as a chronic inflammatory disease of the airways, often in response to inhaled allergens prompting inappropriate activation of the immune response. involving a range of cells including mast cells, Th2 lymphocytes and eosinophils and a wide range of inflammatory mediators. First-line therapy for treatment of persistent asthma involves the use of inhaled corticosteroids (ICS) in combination with inhaled β2-agonists enabling both the control of the underlying airways inflammation and a reduction of airway hyperresponsiveness. However, many patients remain symptomatic despite high-dose therapy. There is therefore a continued unmet clinical need to develop specifically new anti-inflammatory therapies for patients with asthma, either as an add-on therapy to ICS or as replacement monotherapies. The success of fixed dose combination inhalers containing both a bronchodilator and an anti-inflammatory drug has also led to the development of "bifunctional" drugs which are molecules specifically designed to have two distinct pharmacological actions based on distinct pharmacophores. In this review we will discuss these different pharmacological approaches under development for the treatment of bronchial asthma and the available pre-clinical and clinical data.
Collapse
Affiliation(s)
- Richard T Amison
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK -
| | - Clive P Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| |
Collapse
|
17
|
Jin L, Ying ZH, Yu CH, Zhang HH, Yu WY, Wu XN. Isofraxidin ameliorated influenza viral inflammation in rodents via inhibiting platelet aggregation. Int Immunopharmacol 2020; 84:106521. [PMID: 32315950 DOI: 10.1016/j.intimp.2020.106521] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/15/2020] [Accepted: 04/15/2020] [Indexed: 12/12/2022]
Abstract
Platelets have been proved to exacerbate influenza infection and its complications. Inhibition of platelet activation may be a feasible method for preventing severe infection and secondary acute lung injury (ALI). Isofraxidin (IFD) is a natural coumarin isolated from the plants Sarcandra glabra and Siberian ginseng, and exerts anticancer, antioxidant and antiinflammatory effects. In the present study, we examined the therapeutic effects of IFD in ADP- or arachidonic acid (AA)-induced platelet aggregation model and in influenza A virus (IAV)-induced ALI mouse model. The results showed that IFD significantly inhibited platelet aggregation induced by ADP and AA in vitro in a concentration-dependent manner as well as the release of soluble P-selectin and platelet factor 4. Moreover, IFD significantly relieved IAV-induced lung inflammation, reduced the expressions of platelet activation biomarkers (P-selectin and CD61), decreased the serum levels of TNF-α, IL-1β, IL-6 and MIP-2, suppressed peripheral platelet aggregation and prolonged the survival time of infected mice. The western blotting results also demonstrated that IFD reduced the phosphorylation levels of PI3K, AKT and p38 in the activated platelets stimulated by ADP and IAV infection. But IFD did not have any effects on IAV replication. It indicated that IFD ameliorated IAV-induced severe lung damage and lethal infection by suppressing platelet aggregation via regulating PI3K/AKT and MAPK pathways.
Collapse
Affiliation(s)
- Lei Jin
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Zhen-Hua Ying
- Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou 310006, China
| | - Chen-Huan Yu
- Zhejiang Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Huan-Huan Zhang
- Zhejiang Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Wen-Ying Yu
- Zhejiang Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China
| | - Xiao-Ning Wu
- Zhejiang Key Laboratory of Experimental Animal and Safety Evaluation, Hangzhou Medical College, Hangzhou 310013, China.
| |
Collapse
|
18
|
Liu L, Shao Z, Yu H, Zhang W, Wang H, Mei Z. Is the platelet to lymphocyte ratio a promising biomarker to distinguish acute appendicitis? Evidence from a systematic review with meta-analysis. PLoS One 2020; 15:e0233470. [PMID: 32442179 PMCID: PMC7244160 DOI: 10.1371/journal.pone.0233470] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 05/05/2020] [Indexed: 12/29/2022] Open
Abstract
Background Although several previous studies have examined the association between the platelet to lymphocyte ratio (PLR) and acute appendicitis (AA), findings have been controversial. We aimed to systematically assess the available evidence to elucidate the overall relationship between the PLR and AA. Methods Pubmed and Embase databases were searched for all available published literature before August, 2019 by two independent investigators for observational studies reporting the association between the PLR and AA. Random effects models were applied for all meta-analyses. Pooled standardized mean difference (SMD) and 95% confidence interval (CI) were calculated as effect estimates. Results Eleven articles met the inclusion criteria and included in this study. Meta-analysis showed that the level of PLR in the AA group was significantly higher than that in the control group (SMD: 1.19, 95% CI: 0.75 to 1.62, P<0.001). A series of subgroup analyses were conducted to investigate the heterogeneity, showing a significant increase in PLV levels in adults with age ≥30 years (SMD: 1.46, 95% CI: 0.89 to 2.02),compared to those in adult <30 years(SMD: 0.58, 95% CI: 0.12 to 1.04) or in children (SMD: 1.03, 95% CI: 0.51 to 1.56). Compared to non-AA controls, a significant increased PLR level was also observed in non-perforated AA (SMD: 1.23, 95% CI: 0.88 to 1.59) and in AA patients during pregnancy (SMD: 0.70, 95% CI: 0.36 to 1.04), while not in perforated AA (SMD: 2.28, 95% CI: -1.72 to 6.28). Conclusions A significant increase in PLR level is found in patients with AA, indicating that PLR is a promising biomarker for AA. PLR provides a convenient option for emergency department to quickly screen for clinically or radiologically confirmed AA awaiting appendectomy, especially for pregnant women suspected of having AA. More high-quality evidence is needed to further confirm the diagnostic accuracy of PLR for AA.
Collapse
Affiliation(s)
- Lianjie Liu
- Department of Colorectal Surgery, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Zhuo Shao
- Department of General Surgery, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Hang Yu
- Emergency Department, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Wei Zhang
- Department of Colorectal Surgery, Changhai Hospital, Navy Medical University, Shanghai, China
- * E-mail:
| | - Hao Wang
- Department of Colorectal Surgery, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Zubing Mei
- Department of Anorectal Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Anorectal Disease Institute of Shuguang Hospital, Shanghai, China
| |
Collapse
|
19
|
P2Y 12 Inhibition beyond Thrombosis: Effects on Inflammation. Int J Mol Sci 2020; 21:ijms21041391. [PMID: 32092903 PMCID: PMC7073040 DOI: 10.3390/ijms21041391] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/14/2020] [Accepted: 02/15/2020] [Indexed: 12/18/2022] Open
Abstract
The P2Y12 receptor is a key player in platelet activation and a major target for antithrombotic drugs. The beneficial effects of P2Y12 receptor antagonists might, however, not be restricted to the primary and secondary prevention of arterial thrombosis. Indeed, it has been established that platelet activation also has an essential role in inflammation. Additionally, nonplatelet P2Y12 receptors present in immune cells and vascular smooth muscle cells might be effective players in the inflammatory response. This review will investigate the biological and clinical impact of P2Y12 receptor inhibition beyond its platelet-driven antithrombotic effects, focusing on its anti-inflammatory role. We will discuss the potential molecular and cellular mechanisms of P2Y12-mediated inflammation, including cytokine release, platelet–leukocyte interactions and neutrophil extracellular trap formation. Then we will summarize the current evidence on the beneficial effects of P2Y12 antagonists during various clinical inflammatory diseases, especially during sepsis, acute lung injury, asthma, atherosclerosis, and cancer.
Collapse
|
20
|
Imoto Y, Kato A, Takabayashi T, Stevens W, Norton JE, Suh LA, Carter RG, Weibman AR, Hulse KE, Harris KE, Peters AT, Grammer LC, Tan BK, Welch K, Shintani-Smith S, Conley DB, Kern RC, Fujieda S, Schleimer RP. Increased thrombin-activatable fibrinolysis inhibitor levels in patients with chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol 2019; 144:1566-1574.e6. [PMID: 31562871 PMCID: PMC6900453 DOI: 10.1016/j.jaci.2019.08.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 07/02/2019] [Accepted: 08/16/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Chronic rhinosinusitis (CRS) is a heterogeneous chronic inflammatory disease subdivided based on the presence or absence of nasal polyps (NPs). Histologic features of chronic rhinosinusitis with nasal polyps (CRSwNP) include inflammatory cell infiltration and excessive fibrin deposition in NPs. Thrombin-activatable fibrinolysis inhibitor (TAFI) is an enzyme that plays an antifibrinolytic role in the body. The significance of TAFI has been documented in patients with chronic inflammatory diseases, including chronic lung disease; however, it has not been evaluated in the pathogenesis of NPs. OBJECTIVE The objective of this study was to evaluate the potential role of TAFI in the pathogenesis of NPs. METHODS Nasal lavage fluid was collected from control subjects and patients with CRS. We measured levels of thrombin/anti-thrombin complex (TATc) and TAFI protein using an ELISA. RESULTS TATc levels in nasal lavage fluid were significantly increased in patients with CRSwNP and patients with chronic rhinosinusitis without nasal polyps (CRSsNP) compared with control subjects, and TAFI levels in nasal lavage fluid were also significantly increased in patients with CRSwNP compared with those in control subjects and patients with CRSsNP. There was a significant correlation between TATc and TAFI levels in nasal lavage fluid. Interestingly, patients with CRS and asthma showed increased TATc and TAFI levels in nasal lavage fluid compared with those in patients with CRS without asthma, especially patients with CRSwNP. CONCLUSIONS Increased TATc and TAFI levels in nasal passages of patients with CRSwNP might participate in fibrin deposition in NPs and might play a role in the pathogenesis of CRSwNP and asthma.
Collapse
Affiliation(s)
- Yoshimasa Imoto
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill; Department of Otorhinolaryngology Head & Neck Surgery, Faculty of Medical Sciences, University of Fukui, Fukui, Japan.
| | - Atsushi Kato
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill; Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Tetsuji Takabayashi
- Department of Otorhinolaryngology Head & Neck Surgery, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Whitney Stevens
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - James E Norton
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Lydia A Suh
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Roderick G Carter
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Ava R Weibman
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Kathryn E Hulse
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Kathleen E Harris
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Anju T Peters
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Leslie C Grammer
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Bruce K Tan
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Kevin Welch
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | | | - David B Conley
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Robert C Kern
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Shigeharu Fujieda
- Department of Otorhinolaryngology Head & Neck Surgery, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Robert P Schleimer
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill
| |
Collapse
|
21
|
Pitchford S, Cleary S, Arkless K, Amison R. Pharmacological strategies for targeting platelet activation in asthma. Curr Opin Pharmacol 2019; 46:55-64. [PMID: 31026626 DOI: 10.1016/j.coph.2019.03.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/18/2019] [Accepted: 03/22/2019] [Indexed: 12/22/2022]
Abstract
The activation of platelets during host defence and inflammatory disorders has become increasingly documented. Clinical studies of patients with asthma reveal heightened platelet activation and accumulation into lung tissue. Accompanying studies in animal models of allergic lung inflammation, using protocols of experimentally induced thrombocytopenia proclaim an important role for platelets during the leukocyte recruitment cascade, tissue integrity, and lung function. The functions of platelets during these inflammatory events are clearly distinct to platelet functions during haemostasis and clot formation, and have led to the concept that a dichotomy (or polytomy, depending on what else platelets do) in platelet activation exists. The platelet, therefore, presents us with novel opportunities for modulating these inflammatory responses. This review discusses the rationale and effectiveness of current anti-platelet drugs in their use to supress inflammation with regard to asthma, and the need to consider novel possibilities for pharmacological modulation of platelet function associated with inflammation that are pharmacologically distinct to current anti-platelet therapies.
Collapse
Affiliation(s)
- Simon Pitchford
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, UK.
| | - Simon Cleary
- University of California San Francisco (UCSF), Department of Medicine, San Francisco, USA
| | - Kate Arkless
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, UK
| | - Richard Amison
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, UK
| |
Collapse
|
22
|
Sethi GS, Sharma S, Naura AS. PARP inhibition by olaparib alleviates chronic asthma-associated remodeling features via modulating inflammasome signaling in mice. IUBMB Life 2019; 71:1003-1013. [PMID: 30964965 DOI: 10.1002/iub.2048] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 03/29/2019] [Indexed: 01/09/2023]
Abstract
Despite the reported role of poly(ADP-ribose) polymerase (PARP) in asthma inflammation, its contribution during remodeling is not clearly known. The main aim of the current investigation was to examine the potential of olaparib, a pharmacological inhibitor of PARP against airway remodeling using an ovalbumin (OVA)-based murine model of chronic asthma. The results demonstrated that post-challenge olaparib treatment (5 mg/kg i.p., 30 min after OVA exposure) for six weeks (3 days/week) attenuates inflammation, mucus production, and collagen deposition in lungs. Additionally, olaparib blunted the protein expression of STAT-6 and GATA-3 considerably along with a modest reduction in p65-NF-κB phosphorylation. Furthermore, olaparib normalized the OVA-induced redox imbalance as reflected by data on reactive oxygen species, malondialdehyde, protein carbonyls, and reduced glutathione/oxidized glutathione ratio. Interestingly, the protection offered by olaparib was further linked with the altered level of NLRP3 inflammasome-mediated IL-1β release and consequent expression of its downstream targets matrix metalloproteinase-9 and transforming growth factor beta. Suppressed collagen deposition in the lungs correlates well with the reduced expression of vimentin upon olaparib treatment. Finally, olaparib restored the expression of histone deacetylase 2, a steroid-responsive element in asthma. Overall, results suggest that olaparib prevents OVA-induced airway inflammation as well as remodeling via modulating inflammasome signaling in mice. © 2019 IUBMB Life, 1-11, 2019.
Collapse
Affiliation(s)
- Gurupreet S Sethi
- Department of Biochemistry, Panjab University, Chandigarh, Punjab, India
| | - Sukriti Sharma
- Department of Biochemistry, Panjab University, Chandigarh, Punjab, India
| | - Amarjit S Naura
- Department of Biochemistry, Panjab University, Chandigarh, Punjab, India
| |
Collapse
|
23
|
Trinh HKT, Nguyen TVT, Choi Y, Park HS, Shin YS. The synergistic effects of clopidogrel with montelukast may be beneficial for asthma treatment. J Cell Mol Med 2019; 23:3441-3450. [PMID: 30905080 PMCID: PMC6484307 DOI: 10.1111/jcmm.14239] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/30/2019] [Accepted: 02/05/2019] [Indexed: 12/17/2022] Open
Abstract
Platelets modulate asthma pathogenesis by forming the platelet‐eosinophil aggregation (PEA), which facilitates the activation of eosinophils. Platelets exhibit the purinergic receptor (P2Y12R), which responds to cysteinyl leukotriene E4 (LTE4). We have suggested that the combination of an antiplatelet drug (clopidogrel, [Clo]) and montelukast (Mon) would synergistically suppress asthma. BALB/c mice were intraperitoneally sensitized with ovalbumin (OVA) on days 0 and 14 and subsequently challenged on days 28‐30 and 42‐44. Mice were administered with Clo (10 mg/kg), Mon (10 mg/kg) or both drugs (Clo/Mon) orally 30 minutes before the OVA (1%) challenge on days 42‐44. Mice were assayed for airway hyper‐responsiveness (AHR) to methacholine and airway inflammation. Clopidogrel and montelukast attenuated the increased AHR; the combined treatment was more effective than a single treatment for total and eosinophil counts (all P < 0.05). Levels of interleukin (IL)‐4, IL‐5, IL‐13, platelet factor 4, eosinophil peroxidase and LTE4 increased in the bronchoalveolar lavage fluid of asthmatic mice, but these levels decreased in mice treated with Clo/Mon (all P < 0.05). Goblet cell hyperplasia decreased in response to Clo/Mon. Mouse platelets and eosinophils were isolated and co‐cultured for an in vitro assay with 10 µmol/L adenosine diphosphate (ADP), LTE4 (200 nmol/L), Mon (1 µmol/L), Clo (1 µmol/L) and Clo/Mon (1 µmol/L). Flow cytometry revealed that the increased formation of the PEA (%) was fully mediated by ADP and partly mediated by LTE4. Clo/Mon reduced ADP‐induced PEA formation and P‐selectin expression (P < 0.05). In conclusion, Clo/Mon synergistically relieved asthma by inhibiting ADP‐mediated PEA formation.
Collapse
Affiliation(s)
- Hoang Kim Tu Trinh
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea
| | - Thuy Van Thao Nguyen
- Department of Pediatrics, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Youngwoo Choi
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea.,Department of Biomedical Science, Ajou University School of Medicine, Suwon, South Korea
| | - Yoo Seob Shin
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea
| |
Collapse
|