1
|
Derbala MK, Abu-Seida AM, El-Metwally AE, Asfour HAE. A novel method for equine embryo transfer from contaminated recipient mares into second healthy recipients for surviving embryos. J Equine Vet Sci 2024; 142:105200. [PMID: 39303873 DOI: 10.1016/j.jevs.2024.105200] [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: 07/14/2024] [Revised: 08/24/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Embryo transfer (ET) is regarded as one of the reproductive methods for genetic improvement initiatives in the horse business. The current study aimed to increase the success rate of ET by re-flushing and re-transferring of embryos from contaminated recipient mares into second healthy recipients. The study involved 25 recipient mares that demonstrated embryonic vesicles inside intraluminal fluid (ILF) on ultrasound scanning 2 ± 0.5 days after ET. Seven recipient mares were left without re-flushing and re-transferring of embryos (control group) and 18 recipient mares (treated group) were subjected to re-flushing and re-transferring of the embryos to second healthy and prepared recipients. The retrieved fluid from treated mares was subjected to microbiological and cytological examinations. The results revealed mixed and single infections, as well as polymorphonuclear neutrophils (PMNs). The total bacterial count and cytology findings revealed that all first recipient mares had endometritis. All control mares showed embryonic death at 14 ± 1.1 days (13-15 days) of embryonic age. In treated group, microscopic evaluation of retrieved embryos revealed a well-developed thick capsule in 12/18 (66.7 %) of them and a normal capsule in 6/18 (33.3 %). The pregnancy rate in well-developed capsule embryos was greater (66.7 %) than in typical capsule embryos (33.3 %). This study illustrates the value of swabbing the uterus of the recipient mares during estrus prior to ET and their checking at 2 days after ET. In addition to boosting pregnancy rates in ET commercial programs, our novel method can protect the embryos from the harmful effects of the inflammatory process and help them survive.
Collapse
Affiliation(s)
- M K Derbala
- Animal Reproduction Research Institute (ARRI), Agriculture Research Center (ARC). 5 - Hadyek EL-Behoth St. Haram, PO: 12556, Giza, Egypt
| | - A M Abu-Seida
- Department of Surgery, Anesthesiology and Radiology, Faculty of Veterinary Medicine, Cairo University. Giza, PO: 12211, Egypt.
| | - A E El-Metwally
- Pathology Department, Animal Reproduction Research Institute (ARRI), Agriculture Research Center (ARC). Giza, Egypt
| | - H A E Asfour
- Mastitis and Neonatal Diseases Department, Animal Reproduction Research Institute (ARRI), Agriculture Research Center (ARC), Giza, Egypt
| |
Collapse
|
2
|
Li Z, Lu Q. The role of neutrophils in autoimmune diseases. Clin Immunol 2024; 266:110334. [PMID: 39098706 DOI: 10.1016/j.clim.2024.110334] [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: 07/03/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 08/06/2024]
Abstract
Historically, neutrophils have been primarily regarded as short-lived immune cells that act as initial responders to antibacterial immunity by swiftly neutralizing pathogens and facilitating the activation of adaptive immunity. However, recent evidence indicates that their roles are considerably more complex than previously recognized. Neutrophils comprise distinct subpopulations and can interact with various immune cells, release granular proteins, and form neutrophil extracellular traps. These functions are increasingly recognized as contributing factors to tissue damage in autoimmune diseases. This review comprehensively examines the physiological functions and heterogeneity of neutrophils, their interactions with other immune cells, and their significance in autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, antiphospholipid syndrome, antineutrophil cytoplasmic antibody-associated vasculitis, multiple sclerosis, and others. This review aims to provide a deeper understanding of the function of neutrophils in the development and progression of autoimmune disorders.
Collapse
Affiliation(s)
- Zhuoshu Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences &Peking Union Medical College, Nanjing, China; Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Qianjin Lu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences &Peking Union Medical College, Nanjing, China; Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
| |
Collapse
|
3
|
Dai Y, Junho CVC, Schieren L, Wollenhaupt J, Sluimer JC, van der Vorst EPC, Noels H. Cellular metabolism changes in atherosclerosis and the impact of comorbidities. Front Cell Dev Biol 2024; 12:1446964. [PMID: 39188527 PMCID: PMC11345199 DOI: 10.3389/fcell.2024.1446964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/17/2024] [Indexed: 08/28/2024] Open
Abstract
Cell activation and nutrient dysregulation are common consequences of atherosclerosis and its preceding risk factors, such as hypertension, dyslipidemia, and diabetes. These diseases may also impact cellular metabolism and consequently cell function, and the other way around, altered cellular metabolism can impact disease development and progression through altered cell function. Understanding the contribution of altered cellular metabolism to atherosclerosis and how cellular metabolism may be altered by co-morbidities and atherosclerosis risk factors could support the development of novel strategies to lower the risk of CVD. Therefore, we briefly review disease pathogenesis and the principles of cell metabolic pathways, before detailing changes in cellular metabolism in the context of atherosclerosis and comorbidities. In the hypoxic, inflammatory and hyperlipidemic milieu of the atherosclerotic plaque riddled with oxidative stress, metabolism shifts to increase anaerobic glycolysis, the pentose-phosphate pathway and amino acid use. We elaborate on metabolic changes for macrophages, neutrophils, vascular endothelial cells, vascular smooth muscle cells and lymphocytes in the context of atherosclerosis and its co-morbidities hypertension, dyslipidemia, and diabetes. Since causal relationships of specific key genes in a metabolic pathway can be cell type-specific and comorbidity-dependent, the impact of cell-specific metabolic changes must be thoroughly explored in vivo, with a focus on also systemic effects. When cell-specific treatments become feasible, this information will be crucial for determining the best metabolic intervention to improve atherosclerosis and its interplay with co-morbidities.
Collapse
Affiliation(s)
- Yusang Dai
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
- Physical Examination Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Carolina Victoria Cruz Junho
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
| | - Luisa Schieren
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
| | - Julia Wollenhaupt
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
| | - Judith C. Sluimer
- Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Cardiorenal Disease (AMICARE), RWTH Aachen Campus, Aachen, Germany
- Interdisciplinary Centre for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Cardiorenal Disease (AMICARE), RWTH Aachen Campus, Aachen, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| |
Collapse
|
4
|
Yan Q, Zhang X, Xie Y, Yang J, Liu C, Zhang M, Zheng W, Lin X, Huang HT, Liu X, Jiang Y, Zhan SF, Huang X. Bronchial epithelial transcriptomics and experimental validation reveal asthma severity-related neutrophilc signatures and potential treatments. Commun Biol 2024; 7:181. [PMID: 38351296 PMCID: PMC10864370 DOI: 10.1038/s42003-024-05837-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 01/19/2024] [Indexed: 02/16/2024] Open
Abstract
Airway epithelial transcriptome analysis of asthma patients with different severity was used to disentangle the immune infiltration mechanisms affecting asthma exacerbation, which may be advantageous to asthma treatment. Here we introduce various bioinformatics methods and develop two models: an OVA/CFA-induced neutrophil asthma mouse model and an LPS-induced human bronchial epithelial cell damage model. Our objective is to investigate the molecular mechanisms, potential targets, and therapeutic strategies associated with asthma severity. Multiple bioinformatics methods identify meaningful differences in the degree of neutrophil infiltration in asthma patients with different severity. Then, PTPRC, TLR2, MMP9, FCGR3B, TYROBP, CXCR1, S100A12, FPR1, CCR1 and CXCR2 are identified as the hub genes. Furthermore, the mRNA expression of 10 hub genes is determined in vivo and in vitro models. Reperixin is identified as a pivotal drug targeting CXCR1, CXCR2 and MMP9. We further test the potential efficiency of Reperixin in 16HBE cells, and conclude that Reperixin can attenuate LPS-induced cellular damage and inhibit the expression of them. In this study, we successfully identify and validate several neutrophilic signatures and targets associated with asthma severity. Notably, Reperixin displays the ability to target CXCR1, CXCR2, and MMP9, suggesting its potential therapeutic value for managing deteriorating asthma.
Collapse
Affiliation(s)
- Qian Yan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Xinxin Zhang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Yi Xie
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jing Yang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Chengxin Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Miaofen Zhang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Wenjiang Zheng
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xueying Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hui-Ting Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaohong Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Jiang
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China.
| | - Shao-Feng Zhan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Xiufang Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China.
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
5
|
Guo J, Huang Y, Pang L, Zhou Y, Yuan J, Zhou B, Fu M. Association of systemic inflammatory response index with ST segment elevation myocardial infarction and degree of coronary stenosis: a cross-sectional study. BMC Cardiovasc Disord 2024; 24:98. [PMID: 38336634 PMCID: PMC10858502 DOI: 10.1186/s12872-024-03751-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Systemic Inflammatory Response Index (SIRI), a composite inflammatory marker encompassing neutrophils, monocytes, and lymphocytes, has been recognized as a reliable marker of systemic inflammation. This article undertakes an analysis of clinical data from ST-segment Elevation Myocardial Infarction (STEMI) patients, aiming to comprehensively assess the relationship between SIRI, STEMI, and the degree of coronary stenosis. METHODS The study involved 1809 patients diagnosed with STEMI between the years 2020 and 2023. Univariate and multivariate logistic regression analyses were conducted to evaluate the risk factors for STEMI. Receiver operating characteristic (ROC) curves were generated to determine the predictive power of SIRI and neutrophil-to-lymphocyte ratio (NLR). Spearman correlation analysis was performed to assess the correlation between SIRI, NLR, and the Gensini score (GS). RESULTS Multivariate logistic regression analysis showed that the SIRI was the independent risk factor for STEMI (adjusted odds ratio (OR) in the highest quartile = 24.96, 95% confidence interval (CI) = 15.32-40.66, P < 0.001). In addition, there is a high correlation between SIRI and GS (β:28.54, 95% CI: 24.63-32.46, P < 0.001). The ROC curve analysis was performed to evaluate the predictive ability of SIRI and NLR for STEMI patients. The area under the curve (AUC) for SIRI was 0.789. The AUC for NLR was 0.754. Regarding the prediction of STEMI in different gender groups, the AUC for SIRI in the male group was 0.771. The AUC for SIRI in the female group was 0.807. Spearman correlation analysis showed that SIRI exhibited a stronger correlation with GS, while NLR was lower (SIRI: r = 0.350, P < 0.001) (NLR: r = 0.313, P < 0.001). CONCLUSION The study reveals a strong correlation between the SIRI and STEMI as well as the degree of coronary artery stenosis. In comparison to NLR, SIRI shows potential in predicting acute myocardial infarction and the severity of coronary artery stenosis. Additionally, SIRI exhibits a stronger predictive capability for female STEMI patients compared to males.
Collapse
Affiliation(s)
- Jiongchao Guo
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), Hefei, 230000, Anhui, China
| | - Yating Huang
- Department of Endocrinology, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), Hefei, 230000, Anhui, China
| | - Lamei Pang
- Department of Endocrinology, Hefei BOE Hospital, Hefei, 230000, Anhui, China
| | - Yuan Zhou
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), Hefei, 230000, Anhui, China
| | - Jingjing Yuan
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), Hefei, 230000, Anhui, China
| | - Bingfeng Zhou
- Department of Cardiology, Hefei BOE Hospital, Hefei, 230000, Anhui, China.
| | - Minmin Fu
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), Hefei, 230000, Anhui, China.
| |
Collapse
|
6
|
García-Vega D, Sánchez-López D, Rodríguez-Carnero G, Villar-Taibo R, Viñuela JE, Lestegás-Soto A, Seoane-Blanco A, Moure-González M, Bravo SB, Fernández ÁL, González-Juanatey JR, Eiras S. Semaglutide modulates prothrombotic and atherosclerotic mechanisms, associated with epicardial fat, neutrophils and endothelial cells network. Cardiovasc Diabetol 2024; 23:1. [PMID: 38172989 PMCID: PMC10765851 DOI: 10.1186/s12933-023-02096-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Obesity has increased in recent years with consequences on diabetes and other comorbidities. Thus, 1 out of 3 diabetic patients suffers cardiovascular disease (CVD). The network among glucose, immune system, endothelium and epicardial fat has an important role on pro-inflammatory and thrombotic mechanisms of atherogenesis. Since semaglutide, long-acting glucagon like peptide 1- receptor agonist (GLP-1-RA), a glucose-lowering drug, reduces body weight, we aimed to study its effects on human epicardial fat (EAT), aortic endothelial cells and neutrophils as atherogenesis involved-cardiovascular cells. METHODS EAT and subcutaneous fat (SAT) were collected from patients undergoing cardiac surgery. Differential glucose consumption and protein cargo of fat-released exosomes, after semaglutide or/and insulin treatment were analyzed by enzymatic and TripleTOF, respectively. Human neutrophils phenotype and their adhesion to aortic endothelial cells (HAEC) or angiogenesis were analyzed by flow cytometry and functional fluorescence analysis. Immune cells and plasma protein markers were determined by flow cytometry and Luminex-multiplex on patients before and after 6 months treatment with semaglutide. RESULTS GLP-1 receptor was expressed on fat and neutrophils. Differential exosomes-protein cargo was identified on EAT explants after semaglutide treatment. This drug increased secretion of gelsolin, antithrombotic protein, by EAT, modulated CD11b on neutrophils, its migration and endothelial adhesion, induced by adiposity protein, FABP4, or a chemoattractant. Monocytes and neutrophils phenotype and plasma adiposity, stretch, mesothelial, fibrotic, and inflammatory markers on patients underwent semaglutide treatment for 6 months showed a 20% reduction with statistical significance on FABP4 levels and an 80% increase of neutrophils-CD88. CONCLUSION Semaglutide increases endocrine activity of epicardial fat with antithrombotic properties. Moreover, this drug modulates the pro-inflammatory and atherogenic profile induced by the adiposity marker, FABP4, which is also reduced in patients after semaglutide treatment.
Collapse
Affiliation(s)
- David García-Vega
- Cardiology department, Complejo Hospitalario Universitario de Santiago, Travesía de la Choupana SN, 15706, Santiago de Compostela, Spain
- CIBERCV, ISCIII, Madrid, Spain
- University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - David Sánchez-López
- Translational Cardiology, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Gemma Rodríguez-Carnero
- Endocrinology and Nutrition Division, Complejo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
- Epigenomics in Endocrinology and Nutrition Group, Epigenomics Unit, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Rocío Villar-Taibo
- Endocrinology and Nutrition Division, Complejo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
- Neoplasia and Differentiation of Endocrine Cells Group, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Juan E Viñuela
- Translational Cardiology, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Immunology Laboratory, Complejo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Adán Lestegás-Soto
- Translational Cardiology, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Ana Seoane-Blanco
- Cardiology department, Complejo Hospitalario Universitario de Santiago, Travesía de la Choupana SN, 15706, Santiago de Compostela, Spain
- CIBERCV, ISCIII, Madrid, Spain
| | - María Moure-González
- Cardiology department, Complejo Hospitalario Universitario de Santiago, Travesía de la Choupana SN, 15706, Santiago de Compostela, Spain
- CIBERCV, ISCIII, Madrid, Spain
| | - Susana B Bravo
- Proteomics Unit, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ángel L Fernández
- Department of Cardiac Surgery, Complexo Hospitalario Universitario de Santiago, Santiago de Compostela, Spain
| | - José R González-Juanatey
- Cardiology department, Complejo Hospitalario Universitario de Santiago, Travesía de la Choupana SN, 15706, Santiago de Compostela, Spain
- CIBERCV, ISCIII, Madrid, Spain
- University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Sonia Eiras
- Cardiology department, Complejo Hospitalario Universitario de Santiago, Travesía de la Choupana SN, 15706, Santiago de Compostela, Spain.
- CIBERCV, ISCIII, Madrid, Spain.
- Translational Cardiology, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.
| |
Collapse
|
7
|
Christ A, Maas SL, Jin H, Lu C, Legein B, Wijnands E, Temmerman L, Otten J, Isaacs A, Zenke M, Stoll M, Biessen EAL, van der Vorst EPC. In situ lipid-loading activates peripheral dendritic cell subsets characterized by cellular ROS accumulation but compromises their capacity to prime naïve T cells. Free Radic Biol Med 2024; 210:406-415. [PMID: 38061606 DOI: 10.1016/j.freeradbiomed.2023.11.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/22/2023]
Abstract
BACKGROUND AND AIMS Dendritic cells (DCs), professional antigen-presenting cells, play an important role in pathologies by controlling adaptive immune responses. However, their adaptation to and functionality in hypercholesterolemia, a driving factor in disease onset and progression of atherosclerosis remains to be established. METHODS In this study, we addressed the immediate impact of high fat diet-induced hypercholesterolemia in low-density lipoprotein receptor deficient (Ldlr-/-) mice on separate DC subsets, their compartmentalization and functionality. RESULTS While hypercholesterolemia induced a significant rise in bone marrow myeloid and dendritic cell progenitor (MDP) frequency and proliferation rate after high fat diet feeding, it did not affect DC subset numbers in lymphoid tissue. Hypercholesterolemia led to almost immediate and persistent augmentation in granularity of conventional DCs (cDCs), in particular cDC2, reflecting progressive lipid accumulation by these subsets. Plasmacytoid DCs were only marginally and transiently affected. Lipid loading increased co-stimulatory molecule expression and ROS accumulation by cDC2. Despite this hyperactivation, lipid-laden cDC2 displayed a profoundly reduced capacity to stimulate naïve CD4+ T cells. CONCLUSION Our data provide evidence that in hypercholesterolemic conditions, peripheral cDC2 subsets engulf lipids in situ, leading to a more activated status characterized by cellular ROS accumulation while, paradoxically, compromising their T cell priming ability. These findings will have repercussions not only for lipid driven cardiometabolic disorders like atherosclerosis, but also for adaptive immune responses to pathogens and/or endogenous (neo) antigens under conditions of hyperlipidemia.
Collapse
Affiliation(s)
- Anette Christ
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands; Health Office Frankfurt/Main, Frankfurt/Main, Germany.
| | - Sanne L Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany; Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
| | - Han Jin
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Chang Lu
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Bart Legein
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Erwin Wijnands
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Lieve Temmerman
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Jeroen Otten
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands
| | - Aaron Isaacs
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| | - Martin Zenke
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, Aachen, Germany; Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany; Department of Hematology, Oncology and Stem Cell Transplantation, RWTH Aachen University Medical School, 52074, Aachen, Germany
| | - Monika Stoll
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands; Genetic Epidemiology, Institute for Human Genetics, Westfälische Wilhelms-University, Münster, Germany
| | - Erik A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands; Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
| | - Emiel P C van der Vorst
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, Netherlands; Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany; Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany.
| |
Collapse
|
8
|
Kazemi S, Mirzaei R, Karampoor S, Hosseini-Fard SR, Ahmadyousefi Y, Soltanian AR, Keramat F, Saidijam M, Alikhani MY. Circular RNAs in tuberculosis: From mechanism of action to potential diagnostic biomarker. Microb Pathog 2023; 185:106459. [PMID: 37995882 DOI: 10.1016/j.micpath.2023.106459] [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: 05/25/2023] [Revised: 10/01/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (M. tuberculosis), continues to be a major global health concern. Understanding the molecular intricacies of TB pathogenesis is crucial for developing effective diagnostic and therapeutic approaches. Circular RNAs (circRNAs), a class of single-stranded RNA molecules characterized by covalently closed loops, have recently emerged as potential diagnostic biomarkers in various diseases. CircRNAs have been demonstrated to modulate the host's immunological responses against TB, specifically by reducing monocyte apoptosis, augmenting autophagy, and facilitating macrophage polarization. This review comprehensively explores the roles and mechanisms of circRNAs in TB pathogenesis. We also discuss the growing body of evidence supporting their utility as promising diagnostic biomarkers for TB. By bridging the gap between fundamental circRNA biology and TB diagnostics, this review offers insights into the exciting potential of circRNAs in combatting this infectious disease.
Collapse
Affiliation(s)
- Sima Kazemi
- Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Reza Hosseini-Fard
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yaghoub Ahmadyousefi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran; Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Reza Soltanian
- Department of Biostatistics and Epidemiology, School of Public Health, Hamadan University of Medical Sciences, Iran
| | - Fariba Keramat
- Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Massoud Saidijam
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Yousef Alikhani
- Infectious Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
9
|
Riaz B, Sohn S. Neutrophils in Inflammatory Diseases: Unraveling the Impact of Their Derived Molecules and Heterogeneity. Cells 2023; 12:2621. [PMID: 37998356 PMCID: PMC10670008 DOI: 10.3390/cells12222621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023] Open
Abstract
Inflammatory diseases involve numerous disorders and medical conditions defined by an insufficient level of self-tolerance. These diseases evolve over the course of a multi-step process through which environmental variables play a crucial role in the emergence of aberrant innate and adaptive immunological responses. According to experimental data accumulated over the past decade, neutrophils play a significant role as effector cells in innate immunity. However, neutrophils are also involved in the progression of numerous diseases through participation in the onset and maintenance of immune-mediated dysregulation by releasing neutrophil-derived molecules and forming neutrophil extracellular traps, ultimately causing destruction of tissues. Additionally, neutrophils have a wide variety of functional heterogeneity with adverse effects on inflammatory diseases. However, the complicated role of neutrophil biology and its heterogeneity in inflammatory diseases remains unclear. Moreover, neutrophils are considered an intriguing target of interventional therapies due to their multifaceted role in a number of diseases. Several approaches have been developed to therapeutically target neutrophils, involving strategies to improve neutrophil function, with various compounds and inhibitors currently undergoing clinical trials, although challenges and contradictions in the field persist. This review outlines the current literature on roles of neutrophils, neutrophil-derived molecules, and neutrophil heterogeneity in the pathogenesis of autoimmune and inflammatory diseases with potential future therapeutic strategies.
Collapse
Affiliation(s)
- Bushra Riaz
- Department of Biomedical Science, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
| | - Seonghyang Sohn
- Department of Biomedical Science, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| |
Collapse
|
10
|
Liang F, Peng C, Luo X, Wang L, Huang Y, Yin L, Yue L, Yang J, Zhao X. A single-cell atlas of immunocytes in the spleen of a mouse model of Wiskott-Aldrich syndrome. Cell Immunol 2023; 393-394:104783. [PMID: 37944382 DOI: 10.1016/j.cellimm.2023.104783] [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: 05/25/2023] [Revised: 08/28/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
Wiskott-Aldrich syndrome (WAS) is a disorder characterized by rare X-linked genetic immune deficiency with mutations in the Was gene, which is specifically expressed in hematopoietic cells. The spleen plays a major role in hematopoiesis and red blood cell clearance. However, to date, comprehensive analyses of the spleen in wild-type (WT) and WASp-deficient (WAS-KO) mice, especially at the transcriptome level, have not been reported. In this study, single-cell RNA sequencing (scRNA-seq) was adopted to identify various types of immune cells and investigate the mechanisms underlying immune deficiency. We identified 30 clusters and 10 major cell subtypes among 11,269 cells; these cell types included B cells, T cells, dendritic cells (DCs), natural killer (NK) cells, monocytes, macrophages, granulocytes, stem cells and erythrocytes. Moreover, we evaluated gene expression differences among cell subtypes, identified differentially expressed genes (DEGs), and performed enrichment analyses to identify the reasons for the dysfunction in these different cell populations in WAS. Furthermore, some key genes were identified based on a comparison of the DEGs in each cell type involved in specific and nonspecific immune responses, and further analysis showed that these key genes were previously undiscovered pathology-related genes in WAS-KO mice. In summary, we present a landscape of immune cells in the spleen of WAS-KO mice based on detailed data obtained at single-cell resolution. These unprecedented data revealed the transcriptional characteristics of specific and nonspecific immune cells, and the key genes were identified, laying a foundation for future studies of WAS, especially studies into novel and underexplored mechanisms that may improve gene therapies for WAS.
Collapse
Affiliation(s)
- Fangfang Liang
- Department of Rheumatism and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China; Department of Rheumatism and Immunology, Shenzhen Children's Hospital, Shenzhen, China
| | - Cheng Peng
- Department of Radiology, The Third People's Hospital of Shenzhen, Shenzhen, China
| | - Xianze Luo
- Department of Rheumatism and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Linlin Wang
- Department of Rheumatism and Immunology, Shenzhen Children's Hospital, Shenzhen, China
| | - Yanyan Huang
- Department of Rheumatism and Immunology, Shenzhen Children's Hospital, Shenzhen, China
| | - Le Yin
- Department of Rheumatism and Immunology, Shenzhen Children's Hospital, Shenzhen, China
| | - Luming Yue
- Singleron Biotechnologies, Nanjing, Jiangsu, China
| | - Jun Yang
- Department of Rheumatism and Immunology, Shenzhen Children's Hospital, Shenzhen, China.
| | - Xiaodong Zhao
- Department of Rheumatism and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
11
|
Guo J, Chen M, Hong Y, Huang Y, Zhang H, Zhou Y, Zhou B, Fu M. Comparison of the Predicting Value of Neutrophil to high-Density Lipoprotein Cholesterol Ratio and Monocyte to high-Density Lipoprotein Cholesterol Ratio for in-Hospital Prognosis and Severe Coronary Artery Stenosis in Patients with ST-Segment Elevation Acute Myocardial Infarction Following Percutaneous Coronary Intervention: A Retrospective Study. J Inflamm Res 2023; 16:4541-4557. [PMID: 37868828 PMCID: PMC10588721 DOI: 10.2147/jir.s425663] [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: 06/13/2023] [Accepted: 09/30/2023] [Indexed: 10/24/2023] Open
Abstract
Background Neutrophil to high-density lipoprotein cholesterol ratio (NHR) has demonstrated predictive value for coronary artery disease (CAD). However, few research has been conducted on the predictive capacity of NHR for Major Adverse Cardiovascular Events (MACE) following Percutaneous Coronary Intervention (PCI) or the degree of coronary artery stenosis in hospitalized ST-segment elevation myocardial infarction (STEMI) patients. Methods The study involved 486 patients diagnosed with STEMI between the years 2020 and 2023. Univariate and multivariate logistic regression analyses were conducted to evaluate the risk factors for MACE after PCI and severe coronary artery stenosis during hospitalization. Receiver operating characteristic (ROC) curves were generated to determine predictive power of NHR and MHR. Spearman correlation analysis was performed to assess the correlation between NHR, MHR and the Gensini score (GS). Results Multivariate logistic regression analysis showed that the NHR and MHR were the independent risk factor for MACE during hospitalization in STEMI patients (MHR: the odds ratio (OR)=2.347, 95% confidence interval (CI)=1.082-5.089, P=0.031) (NHR: OR=1.092, 95% CI=1.025-1.165, P=0.004). In addition, NHR was also an independent risk factor for high GS (NHR: OR=1.103, 95% CI=1.047-1.162, P<0.001), and the MHR was not an independent risk factor. The ROC curve analysis was performed to evaluate the predictive ability of NHR and MHR for in-hospital MACE in STEMI patients after primary PCI. The area under the curve (AUC) for NHR was 0.681. The AUC for MHR was 0.672. Regarding the prediction of high GS, the AUC for NHR was 0.649. The AUC for MHR was 0.587. Spearman correlation analysis showed that NHR exhibited stronger correlation with GS, while MHR was lower (NHR: r=0.291, P<0.001) (MHR: r=0.156, P<0.001). Conclusion These findings highlight the potential clinical utility of NHR as a predictive indicator in STEMI patients after PCI during hospitalization, both for MACE events and the degree of coronary artery stenosis.
Collapse
Affiliation(s)
- Jiongchao Guo
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People’s Hospital of Hefei), Hefei, Anhui, 230000, People’s Republic of China
- Graduate School, Anhui Medical University, Hefei, Anhui, 230000, People’s Republic of China
| | - Min Chen
- Department of Cardiology, the Second People’s Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230000, People’s Republic of China
| | - Yu Hong
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People’s Hospital of Hefei), Hefei, Anhui, 230000, People’s Republic of China
- Graduate School, Anhui Medical University, Hefei, Anhui, 230000, People’s Republic of China
| | - Yating Huang
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People’s Hospital of Hefei), Hefei, Anhui, 230000, People’s Republic of China
- Department of Endocrinology department, the Third Affiliated Hospital of Anhui Medical University (The First People’s Hospital of Hefei), Hefei, Anhui, 230000, People’s Republic of China
| | - Haiyan Zhang
- Graduate School, Anhui Medical University, Hefei, Anhui, 230000, People’s Republic of China
| | - Yuan Zhou
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People’s Hospital of Hefei), Hefei, Anhui, 230000, People’s Republic of China
- Graduate School, Anhui Medical University, Hefei, Anhui, 230000, People’s Republic of China
| | - Bingfeng Zhou
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People’s Hospital of Hefei), Hefei, Anhui, 230000, People’s Republic of China
- Department of Cardiology, Hefei BOE Hospital, Hefei, Anhui, 230000, People’s Republic of China
| | - Minmin Fu
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People’s Hospital of Hefei), Hefei, Anhui, 230000, People’s Republic of China
| |
Collapse
|
12
|
Cheng W, Bu X, Xu C, Wen G, Kong F, Pan H, Yang S, Chen S. Higher systemic immune-inflammation index and systemic inflammation response index levels are associated with stroke prevalence in the asthmatic population: a cross-sectional analysis of the NHANES 1999-2018. Front Immunol 2023; 14:1191130. [PMID: 37600830 PMCID: PMC10436559 DOI: 10.3389/fimmu.2023.1191130] [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: 03/21/2023] [Accepted: 07/21/2023] [Indexed: 08/22/2023] Open
Abstract
Background Significant evidence suggests that asthma might originate from low-grade systemic inflammation. Previous studies have established a positive association between the systemic immune-inflammation index (SII) and the systemic inflammation response index (SIRI) levels and the risk of stroke. However, it remains unclear whether SII, SIRI and the prevalence of stroke are related in individuals with asthma. Methods The present cross-sectional study used data from the National Health and Nutrition Examination Survey (NHANES) conducted between 1999 and 2018. SII was calculated using the following formula: (platelet count × neutrophil count)/lymphocyte count. SIRI was calculated using the following formula: (neutrophil count × monocyte count)/lymphocyte count. The Spearman rank correlation coefficient was used to determine any correlation between SII, SIRI, and the baseline characteristics. Survey-weighted logistic regression was employed to calculate odds ratios (ORs) and 95% confidence intervals (CIs) to determine the association between SII, SIRI, and stroke prevalence. The predictive value of SII and SIRI for stroke prevalence was assessed through receiver operating characteristic (ROC) curve analysis, with the area under the ROC curve (AUC) being indicative of its predictive value. Additionally, clinical models including SIRI, coronary heart disease, hypertension, age, and poverty income ratio were constructed to evaluate their clinical applicability. Results Between 1999 and 2018, 5,907 NHANES participants with asthma were identified, of which 199 participants experienced a stroke, while the remaining 5,708 participants had not. Spearman rank correlation analysis indicated that neither SII nor SIRI levels exhibited any significant correlation with the baseline characteristics of the participants (r<0.1). ROC curves were used to determine the optimal cut-off values for SII and SIRI levels to classify participants into low- and high-level groups. Higher SII and SIRI levels were associated with a higher prevalence of stroke, with ORs of 1.80 (95% CI, 1.18-2.76) and 2.23 (95% CI, 1.39-3.57), respectively. The predictive value of SIRI (AUC=0.618) for stroke prevalence was superior to that of SII (AUC=0.552). Furthermore, the clinical model demonstrated good predictive value (AUC=0.825), with a sensitivity of 67.1% and specificity of 87.7%. Conclusion In asthmatics, higher levels of SII and SIRI significantly increased the prevalence of stroke, with its association being more pronounced in individuals with coexisting obesity and hyperlipidaemia. SII and SIRI are relatively stable novel inflammatory markers in the asthmatic population, with SIRI having a better predictive value for stroke prevalence than SII.
Collapse
Affiliation(s)
- Wenke Cheng
- Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Xiancong Bu
- Department of Neurology, Zaozhuang Municipal Hospital, Shandong, China
| | - Chunhua Xu
- Department of Recuperation, Lintong Rehabilitation and Recuperation Center, Shanxi, China
| | - Grace Wen
- University Medical Center of Göttingen, Georg-August University, Göttingen, Germany
| | - Fanliang Kong
- University Medical Center of Göttingen, Georg-August University, Göttingen, Germany
| | - Huachun Pan
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shumin Yang
- State Key Laboratory of Agriculture Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Siwei Chen
- Department of Cardiovascular Medicine, Nanchang People's Hospital (The Third Hospital of Nanchang), Jiangxi, China
| |
Collapse
|
13
|
Wang Y, Wan R, Peng W, Zhao X, Bai W, Hu C. Quercetin alleviates ferroptosis accompanied by reducing M1 macrophage polarization during neutrophilic airway inflammation. Eur J Pharmacol 2022; 938:175407. [PMID: 36417973 DOI: 10.1016/j.ejphar.2022.175407] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022]
Abstract
Ferroptosis is a kind of regulated cell death, supporting the pathological process of lung inflammation, including asthma. Quercetin (QCT), a kind of natural dietary flavonoid, exerts anti-inflammatory and anti-ferroptosis effects in various diseases. However, the role of QCT in ferroptosis-associated airway inflammation of neutrophilic asthma remains to be described. Our study aimed to investigate the therapeutic effects of QCT on neutrophilic airway inflammation of asthma. Ferrostatin-1 (Fer-1), as a kind of ferroptosis inhibitor, was used to demonstrate whether neutrophilic airway inflammation of asthma relied on ferroptosis. In our study, the alleviation effect of QCT on neutrophilic airway inflammation was similar to Fer-1. Moreover, the significantly decreased levels of ferroptosis anti-oxidant protein (GPX4 and SLC7A11), increased malondialdehyde (MDA) levels, upregulated levels of 4-hydroxynonenal (4-HNE) expression by immunohistochemistry, and distorted mitochondria morphological changes in the lung tissues suggested lung ferroptosis in neutrophilic airway inflammation, which could be reversed by QCT treatment. In vitro experiments showed that QCT reduced LPS-induced ferroptosis through upregulating cell viability and levels of ferroptosis anti-oxidant protein (SLC7A11 and GPX4), reducing inflammatory cytokines, and decreasing the levels of MDA. Furthermore, ferroptosis was accompanied by enhancing M1 phenotype in neutrophilic airway inflammation, and QCT suppressed ferroptosis by inhibiting the pro-inflammatory M1 profile in vitro and in vivo, just as Fer-1 did. In conclusion, our study found that QCT ameliorated ferroptosis-associated neutrophilic airway inflammation accompanied by inhibiting M1 macrophage polarization. QCT may be a promising ferroptosis inhibitor for neutrophilic airway inflammation.
Collapse
Affiliation(s)
- Yang Wang
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China
| | - Rongjun Wan
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China
| | - Wang Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China
| | - Xincheng Zhao
- Xiangya School of Medicine, Central South University, Hunan, 410008, PR China
| | - Wenxuan Bai
- Xiangya School of Medicine, Central South University, Hunan, 410008, PR China
| | - Chengping Hu
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China.
| |
Collapse
|
14
|
Yamamoto Y, Kadoya K, Terkawi MA, Endo T, Konno K, Watanabe M, Ichihara S, Hara A, Kaneko K, Iwasaki N, Ishijima M. Neutrophils delay repair process in Wallerian degeneration by releasing NETs outside the parenchyma. Life Sci Alliance 2022; 5:e202201399. [PMID: 35961782 PMCID: PMC9375156 DOI: 10.26508/lsa.202201399] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 12/05/2022] Open
Abstract
Although inflammation is indispensable for the repair process in Wallerian degeneration (WD), the role of neutrophils in the WD repair process remains unclear. After peripheral nerve injury, neutrophils accumulate at the epineurium but not the parenchyma in the WD region because of the blood-nerve barrier. An increase or decrease in the number of neutrophils delayed or promoted macrophage infiltration from the epineurium into the parenchyma and the repair process in WD. Abundant neutrophil extracellular traps (NETs) were formed around neutrophils, and its inhibition dramatically increased macrophage infiltration into the parenchyma. Furthermore, inhibition of either MIF or its receptor, CXCR4, in neutrophils decreased NET formation, resulting in enhanced macrophage infiltration into the parenchyma. Moreover, inhibiting MIF for just 2 h after peripheral nerve injury promoted the repair process. These findings indicate that neutrophils delay the repair process in WD from outside the parenchyma by inhibiting macrophage infiltration via NET formation and that neutrophils, NETs, MIF, and CXCR4 are therapeutic targets for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Yasuhiro Yamamoto
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Department of Medicine for Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ken Kadoya
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mohamad Alaa Terkawi
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takeshi Endo
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kohtarou Konno
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Satoshi Ichihara
- Department of Orthopaedic Surgery, Juntendo University Urayasu Hospital, Urayasu, Japan
| | - Akira Hara
- Department of Orthopaedic Surgery, Juntendo University Urayasu Hospital, Urayasu, Japan
| | - Kazuo Kaneko
- Department of Medicine for Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Muneaki Ishijima
- Department of Medicine for Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
15
|
Fan Y, Murgia M, Linder MI, Mizoguchi Y, Wang C, Łyszkiewicz M, Ziȩtara N, Liu Y, Frenz S, Sciuccati G, Partida-Gaytan A, Alizadeh Z, Rezaei N, Rehling P, Dennerlein S, Mann M, Klein C. HAX1-dependent control of mitochondrial proteostasis governs neutrophil granulocyte differentiation. J Clin Invest 2022; 132:153153. [PMID: 35499078 PMCID: PMC9057593 DOI: 10.1172/jci153153] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 03/10/2022] [Indexed: 01/18/2023] Open
Abstract
The relevance of molecular mechanisms governing mitochondrial proteostasis to the differentiation and function of hematopoietic and immune cells is largely elusive. Through dissection of the network of proteins related to HCLS1-associated protein X-1, we defined a potentially novel functional CLPB/HAX1/(PRKD2)/HSP27 axis with critical importance for the differentiation of neutrophil granulocytes and, thus, elucidated molecular and metabolic mechanisms underlying congenital neutropenia in patients with HAX1 deficiency as well as bi- and monoallelic mutations in CLPB. As shown by stable isotope labeling by amino acids in cell culture (SILAC) proteomics, CLPB and HAX1 control the balance of mitochondrial protein synthesis and persistence crucial for proper mitochondrial function. Impaired mitochondrial protein dynamics are associated with decreased abundance of the serine-threonine kinase PRKD2 and HSP27 phosphorylated on serines 78 and 82. Cellular defects in HAX1–/– cells can be functionally reconstituted by HSP27. Thus, mitochondrial proteostasis emerges as a critical molecular and metabolic mechanism governing the differentiation and function of neutrophil granulocytes.
Collapse
Affiliation(s)
- Yanxin Fan
- Department of Pediatrics, Dr. von Hauner Children’s Hospital and Gene Center, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Marta Murgia
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Monika I. Linder
- Department of Pediatrics, Dr. von Hauner Children’s Hospital and Gene Center, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Yoko Mizoguchi
- Department of Pediatrics, Dr. von Hauner Children’s Hospital and Gene Center, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Cong Wang
- Department of Cellular Biochemistry, University Medical Center Goettingen, Goettingen, Germany
| | - Marcin Łyszkiewicz
- Department of Pediatrics, Dr. von Hauner Children’s Hospital and Gene Center, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Natalia Ziȩtara
- Department of Pediatrics, Dr. von Hauner Children’s Hospital and Gene Center, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Yanshan Liu
- Department of Pediatrics, Dr. von Hauner Children’s Hospital and Gene Center, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Stephanie Frenz
- Department of Pediatrics, Dr. von Hauner Children’s Hospital and Gene Center, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Gabriela Sciuccati
- Hematology and Oncology Department, Hospital de Pediatria “Prof. Dr. J.P. Garrahan,” Buenos Aires, Argentina
| | - Armando Partida-Gaytan
- Unidad de Investigación en Inmunodeficiencias Primarias, Instituto Nacional de Pediatría, Mexico City, Mexico
| | | | - Nima Rezaei
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Peter Rehling
- Department of Cellular Biochemistry, University Medical Center Goettingen, Goettingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells,” University of Goettingen, Goettingen, Germany
- Max Planck Institute for Biophysical Chemistry, Goettingen, Germany
| | - Sven Dennerlein
- Department of Cellular Biochemistry, University Medical Center Goettingen, Goettingen, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Christoph Klein
- Department of Pediatrics, Dr. von Hauner Children’s Hospital and Gene Center, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| |
Collapse
|
16
|
D’Agostino A, Di Palma T, Cecchini Gualandi S, Boni R. Fluorescence Spectroscopy for the Diagnosis of Endometritis in the Mare. Animals (Basel) 2022; 12:ani12091157. [PMID: 35565583 PMCID: PMC9101999 DOI: 10.3390/ani12091157] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/18/2022] Open
Abstract
By exploiting the PMN property to produce high quantities of oxygen peroxide to neutralize pathogens, the oxygen peroxide content of uterine cells was measured to diagnose endometritis. After preliminary in vitro studies in which endometrial cells from slaughtered mares were mixed with leukocytes from peripheral blood, endometrial samples were collected by uterine flushing from mares before insemination. Staining endometrial cells with H2DCF-DA was combined with hydroethidine to normalize the fluorescence intensity with the cellular content of the sample. Stained cell smears were assumed as the gold standard of endometritis, and based on this assay, the samples were considered positive (C+) and negative (C−) for endometritis. The amount and the turbidity of fluid recovered by uterine flushing were significantly (p < 0.01) higher in C+ than in C−. Moreover, the oxygen peroxide content of the endometrial cells was significantly higher in the C+ than in the C− group (6.31 ± 1.92 vs. 3.12 ± 1.26, p = 0.001). Using the value of 4.4 as the cutoff level of this fluorescence cytology assay, it was found that only one C− sample exceeded the cutoff level (false positives = 7.7%) while three C+ samples showed values below the cutoff level (false negative = 11.5%).
Collapse
Affiliation(s)
- Andrea D’Agostino
- Department of Sciences, University of Basilicata, Campus Macchia Romana, 85100 Potenza, Italy; (A.D.); (S.C.G.)
| | | | - Stefano Cecchini Gualandi
- Department of Sciences, University of Basilicata, Campus Macchia Romana, 85100 Potenza, Italy; (A.D.); (S.C.G.)
| | - Raffaele Boni
- Department of Sciences, University of Basilicata, Campus Macchia Romana, 85100 Potenza, Italy; (A.D.); (S.C.G.)
- Correspondence: ; Tel.: +39-0971-205017
| |
Collapse
|
17
|
Schwäbe FV, Happonen L, Ekestubbe S, Neumann A. Host Defense Peptides LL-37 and Lactoferrin Trigger ET Release from Blood-Derived Circulating Monocytes. Biomedicines 2022; 10:biomedicines10020469. [PMID: 35203676 PMCID: PMC8962388 DOI: 10.3390/biomedicines10020469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/03/2022] [Accepted: 02/14/2022] [Indexed: 01/08/2023] Open
Abstract
Neutrophils are commonly regarded as the first line of immune response during infection or in tissue injury-induced inflammation. The rapid influx of these cells results in the release of host defense proteins (HDPs) or formation of neutrophil extracellular traps (NETs). As a second wave during inflammation or infection, circulating monocytes arrive at the site. Earlier studies showed that HDPs LL-37 and Lactoferrin (LTF) activate monocytes while neutrophil elastase facilitates the formation of extracellular traps (ETs) in monocytes. However, the knowledge about the impact of HDPs on monocytes remains sparse. In the present study, we investigated the effect of LL-37 and LTF on blood-derived CD14+ monocytes. Both HDPs triggered a significant release of TNFα, nucleosomes, and monocyte ETs. Microscopic analysis indicated that ET formation by LL-37 depends on storage-operated calcium entry (SOCE), mitogen-activated protein kinase (MAPK), and ERK1/2, whereas the LTF-mediated ET release is not affected by any of the here used inhibitors. Quantitative proteomics mass spectrometry analysis of the neutrophil granular content (NGC) revealed a high abundance of Lactoferrin. The stimulation of CD14+ monocytes with NGC resulted in a significant secretion of TNFα and nucleosomes, and the formation of monocyte ETs. The findings of this study provide new insight into the complex interaction of HDPs, neutrophils, and monocytes during inflammation.
Collapse
|
18
|
Xie Y, Abel PW, Casale TB, Tu Y. T H17 cells and corticosteroid insensitivity in severe asthma. J Allergy Clin Immunol 2022; 149:467-479. [PMID: 34953791 PMCID: PMC8821175 DOI: 10.1016/j.jaci.2021.12.769] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/30/2021] [Accepted: 12/15/2021] [Indexed: 02/03/2023]
Abstract
Asthma is classically described as having either a type 2 (T2) eosinophilic phenotype or a non-T2 neutrophilic phenotype. T2 asthma usually responds to classical bronchodilation therapy and corticosteroid treatment. Non-T2 neutrophilic asthma is often more severe. Patients with non-T2 asthma or late-onset T2 asthma show poor response to the currently available anti-inflammatory therapies. These therapeutic failures result in increased morbidity and cost associated with asthma and pose a major health care problem. Recent evidence suggests that some non-T2 asthma is associated with elevated TH17 cell immune responses. TH17 cells producing Il-17A and IL-17F are involved in the neutrophilic inflammation and airway remodeling processes in severe asthma and have been suggested to contribute to the development of subsets of corticosteroid-insensitive asthma. This review explores the pathologic role of TH17 cells in corticosteroid insensitivity of severe asthma and potential targets to treat this endotype of asthma.
Collapse
Affiliation(s)
- Yan Xie
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, USA
| | - Peter W. Abel
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, USA
| | - Thomas B. Casale
- Department of Internal Medicine, University of South Florida School of Medicine, Tampa, FL, USA
| | - Yaping Tu
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, USA
| |
Collapse
|
19
|
Camilli C, Hoeh AE, De Rossi G, Moss SE, Greenwood J. LRG1: an emerging player in disease pathogenesis. J Biomed Sci 2022; 29:6. [PMID: 35062948 PMCID: PMC8781713 DOI: 10.1186/s12929-022-00790-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/11/2022] [Indexed: 12/15/2022] Open
Abstract
The secreted glycoprotein leucine-rich α-2 glycoprotein 1 (LRG1) was first described as a key player in pathogenic ocular neovascularization almost a decade ago. Since then, an increasing number of publications have reported the involvement of LRG1 in multiple human conditions including cancer, diabetes, cardiovascular disease, neurological disease, and inflammatory disorders. The purpose of this review is to provide, for the first time, a comprehensive overview of the LRG1 literature considering its role in health and disease. Although LRG1 is constitutively expressed by hepatocytes and neutrophils, Lrg1-/- mice show no overt phenotypic abnormality suggesting that LRG1 is essentially redundant in development and homeostasis. However, emerging data are challenging this view by suggesting a novel role for LRG1 in innate immunity and preservation of tissue integrity. While our understanding of beneficial LRG1 functions in physiology remains limited, a consistent body of evidence shows that, in response to various inflammatory stimuli, LRG1 expression is induced and directly contributes to disease pathogenesis. Its potential role as a biomarker for the diagnosis, prognosis and monitoring of multiple conditions is widely discussed while dissecting the mechanisms underlying LRG1 pathogenic functions. Emphasis is given to the role that LRG1 plays as a vasculopathic factor where it disrupts the cellular interactions normally required for the formation and maintenance of mature vessels, thereby indirectly contributing to the establishment of a highly hypoxic and immunosuppressive microenvironment. In addition, LRG1 has also been reported to affect other cell types (including epithelial, immune, mesenchymal and cancer cells) mostly by modulating the TGFβ signalling pathway in a context-dependent manner. Crucially, animal studies have shown that LRG1 inhibition, through gene deletion or a function-blocking antibody, is sufficient to attenuate disease progression. In view of this, and taking into consideration its role as an upstream modifier of TGFβ signalling, LRG1 is suggested as a potentially important therapeutic target. While further investigations are needed to fill gaps in our current understanding of LRG1 function, the studies reviewed here confirm LRG1 as a pleiotropic and pathogenic signalling molecule providing a strong rationale for its use in the clinic as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Carlotta Camilli
- Institute of Ophthalmology, University College London, London, UK.
| | - Alexandra E Hoeh
- Institute of Ophthalmology, University College London, London, UK
| | - Giulia De Rossi
- Institute of Ophthalmology, University College London, London, UK
| | - Stephen E Moss
- Institute of Ophthalmology, University College London, London, UK
| | - John Greenwood
- Institute of Ophthalmology, University College London, London, UK
| |
Collapse
|
20
|
Gruijs M, Sewnath CAN, Egmond MV. Therapeutic exploitation of neutrophils to fight cancer. Semin Immunol 2021; 57:101581. [PMID: 34922817 DOI: 10.1016/j.smim.2021.101581] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/22/2021] [Accepted: 12/09/2021] [Indexed: 12/18/2022]
Abstract
Antibody-based immunotherapy is a promising strategy in cancer treatment. Antibodies can directly inhibit tumor growth, induce complement-dependent cytotoxicity and induce Fc receptor-mediated elimination of tumor cells by macrophages and natural killer cells. Until now, however, neutrophils have been largely overlooked as potential effector cells, even though they are the most abundant type of immune cells in the circulation. Neutrophils display heterogeneity, especially in the context of cancer. Therefore, their role in cancer is debated. Nevertheless, neutrophils possess natural anti-tumor properties and appropriate stimulation, i.e. specific targeting via antibody therapy, induces potent tumor cell killing, especially via targeting of the immunoglobulin A Fc receptor (FcαRI, CD89). In this review we address the mechanisms of tumor cell killing by neutrophils and the role of neutrophils in induction of anti-tumor immunity. Moreover, possibilities for therapeutic targeting are discussed.
Collapse
Affiliation(s)
- Mandy Gruijs
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam - Amsterdam Institute for Infection and Immunity, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Celine A N Sewnath
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam - Amsterdam Institute for Infection and Immunity, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Marjolein van Egmond
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam - Amsterdam Institute for Infection and Immunity, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, Cancer Center Amsterdam, De Boelelaan 1117, Amsterdam, the Netherlands.
| |
Collapse
|
21
|
Ma Y, Zhang Y, Zhu L. Role of neutrophils in acute viral infection. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:1186-1196. [PMID: 34472718 PMCID: PMC8589350 DOI: 10.1002/iid3.500] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/23/2021] [Accepted: 07/22/2021] [Indexed: 12/13/2022]
Abstract
Neutrophils play multiple roles in acute viral infections. They restrict viral replication and diffusion through phagocytosis, degranulation, respiratory burst, secretion of cytokines, and the release of neutrophil extracellular traps, as well as, activate the adaptive immune response. However, the overactivation of neutrophils may cause tissue damage and lead to poor outcomes. Additionally, some characteristics and functions of neutrophils, such as cell number, lifespan, and antiviral capability, can be influenced while eliminating viruses. This review provides a general description of the protective and pathological roles of neutrophils in acute viral infection.
Collapse
Affiliation(s)
- Yuan Ma
- Institute of Infectious Diseases, Peking University Ditan Teaching Hospital, Beijing, China
| | - Yue Zhang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Liuluan Zhu
- Institute of Infectious Diseases, Peking University Ditan Teaching Hospital, Beijing, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Filtjens J, Roger A, Quatrini L, Wieduwild E, Gouilly J, Hoeffel G, Rossignol R, Daher C, Debroas G, Henri S, Jones CM, Malissen B, Mackay LK, Moqrich A, Carbone FR, Ugolini S. Nociceptive sensory neurons promote CD8 T cell responses to HSV-1 infection. Nat Commun 2021; 12:2936. [PMID: 34006861 PMCID: PMC8131384 DOI: 10.1038/s41467-021-22841-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/30/2021] [Indexed: 12/13/2022] Open
Abstract
Host protection against cutaneous herpes simplex virus 1 (HSV-1) infection relies on the induction of a robust adaptive immune response. Here, we show that Nav1.8+ sensory neurons, which are involved in pain perception, control the magnitude of CD8 T cell priming and expansion in HSV-1-infected mice. The ablation of Nav1.8-expressing sensory neurons is associated with extensive skin lesions characterized by enhanced inflammatory cytokine and chemokine production. Mechanistically, Nav1.8+ sensory neurons are required for the downregulation of neutrophil infiltration in the skin after viral clearance to limit the severity of tissue damage and restore skin homeostasis, as well as for eliciting robust CD8 T cell priming in skin-draining lymph nodes by controlling dendritic cell responses. Collectively, our data reveal an important role for the sensory nervous system in regulating both innate and adaptive immune responses to viral infection, thereby opening up possibilities for new therapeutic strategies.
Collapse
Affiliation(s)
- Jessica Filtjens
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Anais Roger
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Linda Quatrini
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- Department of Immunology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Elisabeth Wieduwild
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Jordi Gouilly
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Guillaume Hoeffel
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Rafaëlle Rossignol
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Clara Daher
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- Université de Paris, CNRS, Institut Cochin, INSERM, CNRS, Paris, France
| | - Guilhaume Debroas
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Sandrine Henri
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Claerwen M Jones
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Bernard Malissen
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Laura K Mackay
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Aziz Moqrich
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de, Marseille, France
| | - Francis R Carbone
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Sophie Ugolini
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France.
| |
Collapse
|
23
|
Assessment of medullary and extramedullary myelopoiesis in cardiovascular diseases. Pharmacol Res 2021; 169:105663. [PMID: 33979688 DOI: 10.1016/j.phrs.2021.105663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/15/2021] [Accepted: 05/04/2021] [Indexed: 11/23/2022]
Abstract
Recruitment of innate immune cells and their accumulation in the arterial wall and infarcted myocardium has been recognized as a central feature of atherosclerosis and cardiac ischemic injury, respectively. In both, steady state and under pathological conditions, majority of these cells have a finite life span and are continuously replenished from haematopoietic stem/progenitor cell pool residing in the bone marrow and extramedullary sites. While having a crucial role in the cardiovascular disease development, proliferation and differentiation of innate immune cells within haematopoietic compartments is greatly affected by the ongoing cardiovascular pathology. In the current review, we summarize key cells, processes and tissue compartments that are involved in myelopoiesis under the steady state, during atherosclerosis development and in myocardial infarction.
Collapse
|
24
|
Catz SD, McLeish KR. Therapeutic targeting of neutrophil exocytosis. J Leukoc Biol 2020; 107:393-408. [PMID: 31990103 PMCID: PMC7044074 DOI: 10.1002/jlb.3ri0120-645r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 12/11/2022] Open
Abstract
Dysregulation of neutrophil activation causes disease in humans. Neither global inhibition of neutrophil functions nor neutrophil depletion provides safe and/or effective therapeutic approaches. The role of neutrophil granule exocytosis in multiple steps leading to recruitment and cell injury led each of our laboratories to develop molecular inhibitors that interfere with specific molecular regulators of secretion. This review summarizes neutrophil granule formation and contents, the role granule cargo plays in neutrophil functional responses and neutrophil-mediated diseases, and the mechanisms of granule release that provide the rationale for development of our exocytosis inhibitors. We present evidence for the inhibition of granule exocytosis in vitro and in vivo by those inhibitors and summarize animal data indicating that inhibition of neutrophil exocytosis is a viable therapeutic strategy.
Collapse
Affiliation(s)
- Sergio D. Catz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Kenneth R. McLeish
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY
| |
Collapse
|
25
|
Won JE, Lee YS, Park JH, Lee JH, Shin YS, Kim CH, Knowles JC, Kim HW. Hierarchical microchanneled scaffolds modulate multiple tissue-regenerative processes of immune-responses, angiogenesis, and stem cell homing. Biomaterials 2020; 227:119548. [DOI: 10.1016/j.biomaterials.2019.119548] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 10/05/2019] [Accepted: 10/14/2019] [Indexed: 12/27/2022]
|
26
|
Curine Inhibits Macrophage Activation and Neutrophil Recruitment in a Mouse Model of Lipopolysaccharide-Induced Inflammation. Toxins (Basel) 2019; 11:toxins11120705. [PMID: 31816942 PMCID: PMC6950719 DOI: 10.3390/toxins11120705] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/20/2019] [Accepted: 10/22/2019] [Indexed: 11/30/2022] Open
Abstract
Curine is a bisbenzylisoquinoline alkaloid (BBA) with anti-allergic, analgesic, and anti-inflammatory properties. Previous studies have demonstrated that this alkaloid is orally active at non-toxic doses. However, the mechanisms underlying its anti-inflammatory effects remain to be elucidated. This work aimed to investigate the effects of curine on macrophage activation and neutrophil recruitment. Using a murine model of lipopolysaccharide (LPS)-induced pleurisy, we demonstrated that curine significantly inhibited the recruitment of neutrophils in association with the inhibition of cytokines tumor necrosis factor (TNF-α), interleukin (IL)-1β, IL-6, monocyte chemotactic protein (CCL2/MCP-1) as well as leukotriene B4 in the pleural lavage of mice. Curine treatment reduced cytokine levels and the expression of iNOS in in vitro cultures of macrophages stimulated with LPS. Treatment with a calcium channel blocker resulted in comparable inhibition of TNF-α and IL-1β production, as well as iNOS expression by macrophages, suggesting that the anti-inflammatory effects of curine may be related to the inhibition of calcium-dependent mechanisms involved in macrophage activation. In conclusion, curine presented anti-inflammatory effects that are associated with inhibition of macrophage activation and neutrophil recruitment by inhibiting the production of inflammatory cytokines, LTB4 and nitric oxide (NO), and possibly by negatively modulating Ca2+ influx.
Collapse
|
27
|
Wang H, Gao M, Li J, Sun J, Wu R, Han D, Tan J, Wang J, Wang B, Zhang L, Dong Y. MMP-9-positive neutrophils are essential for establishing profibrotic microenvironment in the obstructed kidney of UUO mice. Acta Physiol (Oxf) 2019; 227:e13317. [PMID: 31132220 DOI: 10.1111/apha.13317] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 12/21/2022]
Abstract
AIM Matrix metalloproteinase-9 (MMP9) plays a profibrotic role in renal fibrosis. Neutrophils produce MMP9 in many pathologic models. However, the effect of neutrophil on the progression of renal fibrosis and the relationship of MMP9 to the infiltration of neutrophils into the kidney remain unknown. METHODS The surgery of unilateral ureter obstruction (UUO) was performed in male C57BL/6 mice. Kidneys were collected for analyses on days 0, 1, 3, 5 or 7 following surgery. The inflammatory cells were analysed by flow cytometry. The mRNA and protein levels of renal fibrosis factor and inflammatory factor were measured by qRT-PCR, immumofluorescence and western blot analysis. RESULTS In a mouse kidney model of UUO, neutrophil infiltration significantly increased and neutrophil accumulation reached the highest level at 5 days after the injury. In the obstructed kidney, depleting neutrophils decreased the expression of inflammatory factors, inhibited the accumulation of macrophages including type 2 macrophages and suppressed renal fibrosis. Almost all neutrophils produced MMP9 at the early stage of kidney obstruction. MMP9 attracted neutrophils and inflammatory cells because inhibiting MMP9 suppressed the infiltration of neutrophils and other inflammatory cells and reduced renal fibrosis, regardless of using MMP9 neutralizing antibody or MMP9 inhibitor or different intervening periods of days (0-6, 0-3 or 3-6 were applied after kidney obstruction). CONCLUSION MMP9 promotes neutrophil infiltration by increasing the inflammatory level, macrophage accumulation and renal fibrosis in the obstructed kidney. Inhibiting MMP9 or depleting neutrophils in the early stage of acute kidney injury can relieve the progression of kidney fibrosis.
Collapse
Affiliation(s)
- Haidong Wang
- College of Veterinary Medicine China Agricultural University Beijing China
- College of Animal Science and Veterinary Medicine Shanxi Agricultural University Taigu China
| | - Min Gao
- College of Veterinary Medicine China Agricultural University Beijing China
| | - Jiangbo Li
- College of Veterinary Medicine China Agricultural University Beijing China
| | - Jian Sun
- College of Veterinary Medicine China Agricultural University Beijing China
- Animal Husbandry and Veterinary department Beijing Vocational College of Agriculture Beijing China
| | - Ran Wu
- College of Veterinary Medicine China Agricultural University Beijing China
| | - Deping Han
- College of Veterinary Medicine China Agricultural University Beijing China
| | - Jianmei Tan
- College of Veterinary Medicine China Agricultural University Beijing China
| | - Juan Wang
- Department of Nephrology Shanghai General Hosptial, Shanghai Jiaotong University Shanghai China
| | - Bin Wang
- Institute of Nephrology Zhong Da Hospital, Southeast University Nanjing China
| | - Liping Zhang
- College of Animal Science and Veterinary Medicine Shanxi Agricultural University Taigu China
- Nephrology Division, Department of Medicine Baylor College of Medicine Houston Texas
| | - Yanjun Dong
- College of Veterinary Medicine China Agricultural University Beijing China
| |
Collapse
|
28
|
Zhao J, Huangfu C, Chang Z, Grainger AT, Liu Z, Shi W. Atherogenesis in the Carotid Artery with and without Interrupted Blood Flow of Two Hyperlipidemic Mouse Strains. J Vasc Res 2019; 56:241-254. [PMID: 31536996 DOI: 10.1159/000502691] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 08/13/2019] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Atherosclerosis in the carotid arteries is a common cause of ischemic stroke. We examined atherogenesis in the left carotid artery with and without interrupted blood flow of C57BL/6 (B6) and C3H-Apoe-deficient (Apoe-/-) mouse strains. METHODS Blood flow was interrupted by ligating the common carotid artery near its bifurcation in one group of mice and another group was not interrupted. RESULTS Without interference with blood flow, C3H-Apoe-/- mice developed no atherosclerosis in the carotid artery, while B6-Apoe-/- mice formed advanced atherosclerotic lesions (98,019 ± 10,594 μm2/section) after 12 weeks of a Western diet. When blood flow was interrupted by ligating the common carotid artery near its bifurcation, C3H-Apoe-/- mice showed fatty streak lesions 2 weeks after ligation, and by 4 weeks fibrous lesions had formed, although they were smaller than in B6-Apoe-/- mice. Neutrophil adhesion to endothelium and infiltration in lesions was observed in ligated arteries of both strains. Treatment of B6-Apoe-/- mice with antibody against neutrophils had little effect on lesion size. CONCLUSIONS These findings demonstrate the dramatic influences of genetic backgrounds and blood flow on atherogenesis in the carotid artery of hyperlipidemic mice.
Collapse
Affiliation(s)
- Jian Zhao
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, USA.,Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chaoji Huangfu
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, USA.,Center for Disease Control and Prevention, Western Theater Command, Lanzhou, China
| | - Zhihui Chang
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, USA.,Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Andrew T Grainger
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
| | - Zhaoyu Liu
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Weibin Shi
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, USA, .,Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA,
| |
Collapse
|
29
|
Hey YY, O'Neill TJ, O'Neill HC. A novel myeloid cell in murine spleen defined through gene profiling. J Cell Mol Med 2019; 23:5128-5143. [PMID: 31210415 PMCID: PMC6653018 DOI: 10.1111/jcmm.14382] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 04/04/2019] [Accepted: 04/17/2019] [Indexed: 12/17/2022] Open
Abstract
A novel myeloid antigen presenting cell can be generated through in vitro haematopoiesis in long‐term splenic stromal cocultures. The in vivo equivalent subset was recently identified as phenotypically and functionally distinct from the spleen subsets of macrophages, conventional (c) dendritic cells (DC), resident monocytes, inflammatory monocytes and eosinophils. This novel subset which is myeloid on the basis of cell surface phenotype, but dendritic‐like on the basis of cell surface marker expression and antigen presenting function, has been tentatively labelled “L‐DC.” Transcriptome analysis has now been employed to determine the lineage relationship of this cell type with known splenic cDC and monocyte subsets. Principal components analysis showed separation of “L‐DC” and monocytes from cDC subsets in the second principal component. Hierarchical clustering then indicated a close lineage relationship between this novel subset, resident monocytes and inflammatory monocytes. Resident monocytes were the most closely aligned, with no genes specifically expressed by the novel subset. This subset, however, showed upregulation of genes reflecting both dendritic and myeloid lineages, with strong upregulation of several genes, particularly CD300e. While resident monocytes were found to be dependent on Toll‐like receptor signalling for development and were reduced in number in Myd88‐/‐ and Trif‐/‐ mutant mice, both the novel subset and inflammatory monocytes were unaffected. Here, we describe a novel myeloid cell type closely aligned with resident monocytes in terms of lineage but distinct in terms of development and functional capacity.
Collapse
Affiliation(s)
- Ying-Ying Hey
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QLD, Australia
| | | | - Helen C O'Neill
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QLD, Australia
| |
Collapse
|
30
|
Gu Y, Hsu ACY, Pang Z, Pan H, Zuo X, Wang G, Zheng J, Wang F. Role of the Innate Cytokine Storm Induced by the Influenza A Virus. Viral Immunol 2019; 32:244-251. [PMID: 31188076 DOI: 10.1089/vim.2019.0032] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Influenza A viruses (IAVs) can be classified into dozens of subtypes based on their hemagglutinin (HA) and neuraminidase (NA) proteins. To date, 18 HA subtypes and 11 NA subtypes of IAVs that spread in animals and humans have been found. Following infection, the IAV first induces the innate immune system, which can rapidly recruit innate immune cells and cytokines to the site of infection. Influenza-induced cytokine storms have been associated with uncontrolled proinflammatory responses, which may lead to significant immunopathy and severe disease. Cytokine storms are complicated by several types of cytokines and chemokines that have various activities. In addition to their direct effects, their crossregulation causes cytokine networks to form; these networks determine the outcome of viral infections. In this review, we focus on cytokine storms and their signaling pathways that are triggered by the different subtypes of IAV.
Collapse
Affiliation(s)
- Yinuo Gu
- 1Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Alan Chen-Yu Hsu
- 2Priority Research Center for Healthy Lungs, Faculty of Health and Medicine, the University of Newcastle, Newcastle, New South Wales, Australia
| | - Zhiqiang Pang
- 1Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - He Pan
- 1Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xu Zuo
- 1Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Guoqiang Wang
- 1Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jingtong Zheng
- 1Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Fang Wang
- 1Department of Pathogeny Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
31
|
Yan X, Xie B, Wu G, Hu J, Wang D, Cai X, Li J. Interleukin-37: The Effect of Anti-Inflammatory Response in Human Coronary Artery Endothelial Cells. Mediators Inflamm 2019; 2019:2650590. [PMID: 30728750 PMCID: PMC6341264 DOI: 10.1155/2019/2650590] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/07/2018] [Accepted: 11/06/2018] [Indexed: 02/05/2023] Open
Abstract
Interleukin-37 (IL-37) is unique in the IL-1 family since it broadly suppresses innate immunity and elevates in humans with inflammatory and autoimmune diseases. IL-37 shows definite groups and transcripts for human IL37 gene, but it is still not completely understood the effect and mechanisms of inflammatory response in endothelial cells. It is well accepted that endothelial dysfunction caused by inflammation is a key initiating event in atherosclerotic plaque formation, which leads to the occurrence and development of the cardiovascular adverse events in clinical since the inflammatory responses of endothelial cells could induce and enhance the deposition of extensive lipid and the formation of atherosclerotic plaque in the intima. Thus, it is essential to investigate the role and potential mechanisms in endothelial inflammatory response to prevent the formation and development of many cardiovascular diseases including atherosclerosis. So far, the recent studies have revealed that IL-37 is able to inhibit inflammatory response by suppressing the TLR2-NF-κB-ICAM-1 pathway intracellularly in human coronary artery endothelial cells (HCAECs). Further, the role of IL-37 may be related to the IL-18 pathway extracellularly and involved in the adhesion and transmigration of neutrophils in HCAECs.
Collapse
Affiliation(s)
- Xianfeng Yan
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Bin Xie
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Guihai Wu
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Jing Hu
- Department of Cardiology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, China
| | - Di Wang
- Department of Dermatovenereology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Xiangna Cai
- Department of Plastic Surgery, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Jilin Li
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| |
Collapse
|
32
|
Avazi DO, Awasum AC, Hassan AZ, Ayo JO, Aluwong T, Muhammed ST, Simon AY, Suleiman MH, Kudi AC. Evaluation of levels of interleukin-6, interleukin-8 and some haematologic parameters of dogs with cutaneous wounds. Cytokine 2018; 113:128-138. [PMID: 30539777 DOI: 10.1016/j.cyto.2018.06.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/08/2018] [Accepted: 06/18/2018] [Indexed: 11/28/2022]
Abstract
The aim of the study was to investigate the time-course of serum and wound fluids interleukin (IL)-6 and IL-8 levels in dogs with cutaneous wounds and their relationship with some haematologic parameters. The experimental group comprised of six adult dogs that underwent surgery with wounds (n = 6) on the mid lateral aspect of the right antebrachium; and control group of six, apparently, healthy intact (free from cutaneous wounds) adult dogs, comprising equal number of both sexes. Vital signs evaluated were within normal limits. Samples of blood, serum and wound fluids harvested pre- and at 12 h, 36 h, 60 h, 156 h and 324 h post-injury, were utilised for IL-6 and IL-8 assay and haematology. Peak concentrations of IL-6 in wound fluid (1.33. ± 0.33 ng/mL) and serum (0.82 ± 0.24 ng/mL) of the experimental group at 12 h post-operation were higher (P < 0.01) than the control (0.30 ± 0.05 ng/mL). Concentrations of IL-8 at 12 h and 60 h in wound fluid (0.21 ± 0.05 ng/mL and 0.22 ± 0.11 ng/mL) respectively were lower (P < 0.05) than serum (0.71 ± 0.21 ng/mL and 0.73 ± 0.24 ng/mL) respectively in the experimental group and corresponding values recorded in controls (0.34 ± 0.09 ng/mL and 0.36 ± 0.14 ng/mL). The haematological and biochemical parameters exhibited minimum fluctuations, but values were within normal ranges. Significant correlations were obtained between serum and wound fluid IL-6 (r = 0.827, P < 0.05); wound fluid IL-6 and monocyte count (r = 0.818, P < 0.04); wound fluid IL-6 and haematocrit (r = -0.894, P < 0.05). There was a positive correlation between serum IL-8 and serum IL-6 (r = 0.622, P > 0.05) and serum IL-8 and wound fluid IL-8 (r = 0.718, P > 0.05) in the experimental group. In conclusion, IL-6 and IL-8 exerted modulated inflammatory processes following cutaneous wounds in dogs. Further studies are required to investigate the expression patterns of IL-6 and IL-8 in cutaneous wounds in order to improve the quality of management of cutaneous wounds.
Collapse
Affiliation(s)
| | | | | | | | - Tagang Aluwong
- Department of Veterinary Physiology, ABU, Zaria, Nigeria
| | | | | | | | | |
Collapse
|
33
|
de Graauw E, Sitaru C, Horn MP, Borradori L, Yousefi S, Simon D, Simon HU. Monocytes enhance neutrophil-induced blister formation in an ex vivo model of bullous pemphigoid. Allergy 2018; 73:1119-1130. [PMID: 29222810 DOI: 10.1111/all.13376] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Lesions of bullous pemphigoid (BP), an autoimmune subepidermal blistering disease characterized by the presence of tissue-bound and circulating autoantibodies to hemidesmosomal antigens, harbor a mixed inflammatory cellular infiltrate. In various models, neutrophils, eosinophils, mast cells, monocytes as well as B and T cells have been shown to be involved in the pathogenesis of BP. However, their interactions with and effective role in blister formation remain uncertain. This study was aimed at investigating the effect of monocyte/neutrophil interaction on blister formation in an ex vivo BP model. METHODS Skin cryosections were incubated with purified human neutrophils and monocytes, in the presence or absence of BP autoantibodies. Production of reactive oxygen species (ROS), degranulation, mediator release (neutrophil elastase [NE], myeloperoxidase [MPO], matrix metalloproteinase-9 [MMP-9]), binding of Fcγ receptor (CD16, CD32, CD64), and cell adhesion (CD18, ICAM-1) was investigated using appropriate inhibitors. Dermal-epidermal separation (DES) was assessed by light microscopy and quantified by Fiji software. RESULTS Monocytes and neutrophils synergistically interact resulting in a significantly higher DES compared to either monocytes or neutrophils separately (P < .0001). Monocyte/neutrophil-induced DES was associated with increased ROS production and was dependent on adhesion and FcγRIII binding. Upon stimulation by the granule-poor fraction of monocyte supernatants, neutrophils increased their release of MMP-9, thereby also DES at the dermal-epidermal junction of skin cryosections. CONCLUSION Our observations suggest that the interaction of cells, as shown here for monocytes and neutrophils, enhances mediator release resulting in an increased subepidermal blister formation. Thus, blocking intercellular cross talk promises a new therapeutic approach for blocking tissue damage in BP.
Collapse
Affiliation(s)
- E. de Graauw
- Institute of Pharmacology; University of Bern; Bern Switzerland
| | - C. Sitaru
- Department of Dermatology; University Medical Center Freiburg; Freiburg Germany
- BIOSS Centre for Biological Signalling Studies; Albert Ludwig University of Freiburg; Freiburg Germany
| | - M. P. Horn
- University Institute of Clinical Chemistry and Centre of Laboratory Medicine; Inselspital; Bern University Hospital; University of Bern; Bern Switzerland
| | - L. Borradori
- Department of Dermatology, Inselspital; Bern University Hospital; University of Bern; Bern Switzerland
| | - S. Yousefi
- Institute of Pharmacology; University of Bern; Bern Switzerland
| | - D. Simon
- Department of Dermatology, Inselspital; Bern University Hospital; University of Bern; Bern Switzerland
| | - H.-U. Simon
- Institute of Pharmacology; University of Bern; Bern Switzerland
| |
Collapse
|
34
|
Prame Kumar K, Nicholls AJ, Wong CHY. Partners in crime: neutrophils and monocytes/macrophages in inflammation and disease. Cell Tissue Res 2018; 371:551-565. [PMID: 29387942 PMCID: PMC5820413 DOI: 10.1007/s00441-017-2753-2] [Citation(s) in RCA: 254] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 11/21/2017] [Indexed: 02/07/2023]
Abstract
Neutrophils are becoming recognized as highly versatile and sophisticated cells that display de novo synthetic capacity and potentially prolonged lifespan. Emerging concepts such as neutrophil heterogeneity and plasticity have revealed that, under pathological conditions, neutrophils may differentiate into discrete subsets defined by distinct phenotypic and functional characteristics. Indeed, these newly described neutrophil subsets will undoubtedly add to the already complex interactions between neutrophils and other immune cell types for an effective immune response. The interactions between neutrophils and monocytes/macrophages enable the host to efficiently defend against and eliminate foreign pathogens. However, it is also becoming increasingly clear that these interactions can be detrimental to the host if not tightly regulated. In this review, we will explore the functional cooperation of neutrophil and monocytes/macrophages in homeostasis, during acute inflammation and in various disease settings. We will discuss this in the context of cardiovascular disease in the form of atherosclerosis, an autoimmune disease mainly occurring in the kidneys, as well as the unique intestinal immune response of the gut that does not conform to the norms of the typical immune system.
Collapse
Affiliation(s)
- Kathryn Prame Kumar
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, 3168, Australia
| | - Alyce J Nicholls
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, 3168, Australia
| | - Connie H Y Wong
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, 3168, Australia.
| |
Collapse
|
35
|
Anderson MC, Chaze T, Coïc YM, Injarabian L, Jonsson F, Lombion N, Selimoglu-Buet D, Souphron J, Ridley C, Vonaesch P, Baron B, Arena ET, Tinevez JY, Nigro G, Nothelfer K, Solary E, Lapierre V, Lazure T, Matondo M, Thornton D, Sansonetti PJ, Baleux F, Marteyn BS. MUB 40 Binds to Lactoferrin and Stands as a Specific Neutrophil Marker. Cell Chem Biol 2018; 25:483-493.e9. [PMID: 29478905 DOI: 10.1016/j.chembiol.2018.01.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/04/2017] [Accepted: 01/26/2018] [Indexed: 02/07/2023]
Abstract
Neutrophils represent the most abundant immune cells recruited to inflamed tissues. A lack of dedicated tools has hampered their detection and study. We show that a synthesized peptide, MUB40, binds to lactoferrin, the most abundant protein stored in neutrophil-specific and tertiary granules. Lactoferrin is specifically produced by neutrophils among other leukocytes, making MUB40 a specific neutrophil marker. Naive mammalian neutrophils (human, guinea pig, mouse, rabbit) were labeled by fluorescent MUB40 conjugates (-Cy5, Dylight405). A peptidase-resistant retro-inverso MUB40 (RI-MUB40) was synthesized and its lactoferrin-binding property validated. Neutrophil lactoferrin secretion during in vitro Shigella infection was assessed with RI-MUB40-Cy5 using live cell microscopy. Systemically administered RI-MUB40-Cy5 accumulated at sites of inflammation in a mouse arthritis inflammation model in vivo and showed usefulness as a potential tool for inflammation detection using non-invasive imaging. Improving neutrophil detection with the universal and specific MUB40 marker will aid the study of broad ranges of inflammatory diseases.
Collapse
Affiliation(s)
- Mark C Anderson
- Institut Pasteur, Unité de Pathogénie Microbienne Moléculaire, 28 rue du Dr Roux, 75724 Paris Cedex 15, France; INSERM Unité 1202, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Thibault Chaze
- Institut Pasteur / CNRS USR 2000 Mass Spectrometry for Biology, Proteomics Platform, CITECH, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Yves-Marie Coïc
- Institut Pasteur, Unité de Chimie des Biomolécules, CNRS UMR 3523, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Louise Injarabian
- Institut Pasteur, Unité de Pathogénie Microbienne Moléculaire, 28 rue du Dr Roux, 75724 Paris Cedex 15, France; INSERM Unité 1202, 28 rue du Dr Roux, 75724 Paris Cedex 15, France; CNRS, IBGC, Cell Energetic Metabolism, 1 rue Camille Saint Saëns CS 61390, 33077 Bordeaux Cedex, France
| | - Friederike Jonsson
- Institut Pasteur, Département d'Immunologie, 25 rue du Docteur Roux, 75024 Paris Cedex 15, France; INSERM Unité 1222, 25 rue du Dr Roux, 75015 Paris Cedex 15, France
| | - Naelle Lombion
- Institut Gustave Roussy, Laboratoire de Thérapie Cellulaire, 114 rue Edouard Vaillant, 94800 Villejuif, France
| | | | - Judith Souphron
- Institut Pasteur, Unité de Pathogénie Microbienne Moléculaire, 28 rue du Dr Roux, 75724 Paris Cedex 15, France; INSERM Unité 1202, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Caroline Ridley
- University of Manchester, Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, A.V. Hill Building, Manchester M13 9PT, UK
| | - Pascale Vonaesch
- Institut Pasteur, Unité de Pathogénie Microbienne Moléculaire, 28 rue du Dr Roux, 75724 Paris Cedex 15, France; INSERM Unité 1202, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Bruno Baron
- Institut Pasteur, Plate-Forme de Biophysique Moléculaire, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Ellen T Arena
- Institut Pasteur, Unité de Pathogénie Microbienne Moléculaire, 28 rue du Dr Roux, 75724 Paris Cedex 15, France; INSERM Unité 1202, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Jean-Yves Tinevez
- Institut Pasteur, CITECH, Imagopole, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Giulia Nigro
- Institut Pasteur, Unité de Pathogénie Microbienne Moléculaire, 28 rue du Dr Roux, 75724 Paris Cedex 15, France; INSERM Unité 1202, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Katharina Nothelfer
- Institut Pasteur, Unité de Pathogénie Microbienne Moléculaire, 28 rue du Dr Roux, 75724 Paris Cedex 15, France; INSERM Unité 1202, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Eric Solary
- Institut Gustave Roussy Inserm U1009, 114 rue Edouard Vaillant, 94800 Villejuif, France
| | - Valérie Lapierre
- Institut Gustave Roussy, Laboratoire de Thérapie Cellulaire, 114 rue Edouard Vaillant, 94800 Villejuif, France
| | - Thierry Lazure
- APHP Hôpital du Kremlin-Bicêtre, 78 rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France
| | - Mariette Matondo
- Institut Pasteur / CNRS USR 2000 Mass Spectrometry for Biology, Proteomics Platform, CITECH, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - David Thornton
- Institut Pasteur, Département d'Immunologie, 25 rue du Docteur Roux, 75024 Paris Cedex 15, France
| | - Philippe J Sansonetti
- Institut Pasteur, Unité de Pathogénie Microbienne Moléculaire, 28 rue du Dr Roux, 75724 Paris Cedex 15, France; INSERM Unité 1202, 28 rue du Dr Roux, 75724 Paris Cedex 15, France; Collège de France, Paris, France
| | - Françoise Baleux
- Institut Pasteur, Unité de Chimie des Biomolécules, CNRS UMR 3523, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Benoit S Marteyn
- Institut Pasteur, Unité de Pathogénie Microbienne Moléculaire, 28 rue du Dr Roux, 75724 Paris Cedex 15, France; INSERM Unité 1202, 28 rue du Dr Roux, 75724 Paris Cedex 15, France; Institut Gustave Roussy, Laboratoire de Thérapie Cellulaire, 114 rue Edouard Vaillant, 94800 Villejuif, France.
| |
Collapse
|
36
|
Kim YS, Ahn Y. Functional Relevance of Macrophage-mediated Inflammation to Cardiac Regeneration. Chonnam Med J 2018; 54:10-16. [PMID: 29399560 PMCID: PMC5794473 DOI: 10.4068/cmj.2018.54.1.10] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/06/2017] [Accepted: 12/19/2017] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular disease remains the leading cause of death worldwide and regenerative medicine is a promising therapeutic option for this disease. We have developed various techniques to attenuate the cardiac remodeling and to regenerate cardiovascular systems via stem cell application. Besides cell therapy, we are interested in the modulation of pathological inflammation mediated by macrophages in the damaged heart tissue to arouse endogenous reparative responses with biocompatible small molecules. Certainly, current understanding of mechanisms of tissue regeneration will lead to the development of innovative regenerative medicine for cardiovascular disease.
Collapse
Affiliation(s)
- Yong Sook Kim
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju, Korea.,Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Youngkeun Ahn
- Cell Regeneration Research Center, Chonnam National University Hospital, Gwangju, Korea.,Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea
| |
Collapse
|
37
|
Goh JG, Ravikumar S, Win MS, Cao Q, Tan AL, Lim JHJ, Leong W, Herbrecht R, Troke PF, Kullberg BJ, Netea MG, Chng WJ, Dan YY, Chai LYA. Neutrophils differentially attenuate immune response to Aspergillus infection through complement receptor 3 and induction of myeloperoxidase. Cell Microbiol 2017; 20. [PMID: 29088499 DOI: 10.1111/cmi.12798] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 10/07/2017] [Accepted: 10/25/2017] [Indexed: 12/11/2022]
Abstract
Invasive aspergillosis (IA) remains a major cause of morbidity in immunocompromised hosts. This is due to the inability of the host immunity to respond appropriately to Aspergillus. An established risk factor for IA is neutropenia that is encountered by patients undergoing chemotherapy. Herein, we investigate the role of neutrophils in modulating host response to Aspergillus. We found that neutrophils had the propensity to suppress proinflammatory cytokine production but through different mechanisms for specific cytokines. Cellular contact was requisite for the modulation of interleukin-1 beta production by Aspergillus with the involvement of complement receptor 3. On the other hand, inhibition of tumour necrosis factor-alpha production (TNF-α) was cell contact-independent and mediated by secreted myeloperoxidase. Specifically, the inhibition of TNF-α by myeloperoxidase was through the TLR4 pathway and involved interference with the mRNA transcription of TNF receptor-associated factor 6/interferon regulatory factor 5. Our study illustrates the extended immune modulatory role of neutrophils beyond its primary phagocytic function. The absence of neutrophils and loss of its inhibitory effect on cytokine production explains the hypercytokinemia seen in neutropenic patients when infected with Aspergillus.
Collapse
Affiliation(s)
- Jessamine G Goh
- Division of Infectious Diseases, University Medicine Cluster, National University Health System, Singapore, Singapore
| | - Sharada Ravikumar
- Division of Infectious Diseases, University Medicine Cluster, National University Health System, Singapore, Singapore
| | - Mar Soe Win
- Division of Infectious Diseases, University Medicine Cluster, National University Health System, Singapore, Singapore.,Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Qiong Cao
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ai Ling Tan
- Department of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Joan H J Lim
- Division of Infectious Diseases, University Medicine Cluster, National University Health System, Singapore, Singapore
| | - Winnie Leong
- Division of Infectious Diseases, University Medicine Cluster, National University Health System, Singapore, Singapore
| | - Raoul Herbrecht
- Department of Oncology and Hematology, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | | | - Bart Jan Kullberg
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Nijmegen, the Netherlands
| | - Wee Joo Chng
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute, National University of Singapore, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yock Young Dan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Division of Gastroenterology and Hepatology, University Medicine Cluster, National University Health System, Singapore, Singapore
| | - Louis Y A Chai
- Division of Infectious Diseases, University Medicine Cluster, National University Health System, Singapore, Singapore.,Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
38
|
Chang Z, Huangfu C, Grainger AT, Zhang J, Guo Q, Shi W. Accelerated atherogenesis in completely ligated common carotid artery of apolipoprotein E-deficient mice. Oncotarget 2017; 8:110289-110299. [PMID: 29299147 PMCID: PMC5746382 DOI: 10.18632/oncotarget.22685] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/05/2017] [Indexed: 12/22/2022] Open
Abstract
Complete ligation of the common carotid artery near its bifurcation induces neointimal formation due to smooth muscle cell proliferation in normolipidemic wild-type mice, but it was unknown what would happen to hyperlipidemic apolipoprotein E-deficient (Apoe-/-) mice. Examination of these mice revealed rapid development of atherosclerotic lesions in completely ligated carotid arteries within 4 weeks. Mice were fed a Western diet, starting 1 week before ligation, or a chow diet. Foam cell lesions formed as early as 1 week after ligation in mice fed the Western diet and 2 weeks in mice fed the chow diet. Fibrous lesions comprised of foam cells and smooth muscle cells and more advance lesions containing neovessels occurred at 2 and 4 weeks after ligation, respectively, in the Western diet group. Lesions were larger and more advanced in the Western diet group than the chow group. Neutrophil infiltration was observed in growing intimal lesions in both diet groups, while CD8+ T cells were found in lesions of chow-fed mice. This study demonstrates that Apoe-/- mice develop the entire spectrum of atherosclerosis in ligated carotid arteries in an accelerated manner and this model could be a valuable tool for investigating the development and therapy of atherosclerosis.
Collapse
Affiliation(s)
- Zhihui Chang
- Department of Radiology & Medical Imaging, University of Virginia, Charlottesville, Virginia, USA
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chaoji Huangfu
- Department of Radiology & Medical Imaging, University of Virginia, Charlottesville, Virginia, USA
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Andrew T. Grainger
- Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
| | - Jingang Zhang
- Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing Institute of Transfusion Medicine, Beijing, China
| | - Qiyong Guo
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Weibin Shi
- Department of Radiology & Medical Imaging, University of Virginia, Charlottesville, Virginia, USA
- Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
39
|
Ray A, Kolls JK. Neutrophilic Inflammation in Asthma and Association with Disease Severity. Trends Immunol 2017; 38:942-954. [PMID: 28784414 PMCID: PMC5711587 DOI: 10.1016/j.it.2017.07.003] [Citation(s) in RCA: 284] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 06/28/2017] [Accepted: 07/11/2017] [Indexed: 01/22/2023]
Abstract
Asthma is a chronic inflammatory disorder of the airways. While the local infiltration of eosinophils and mast cells, and their role in the disease have long been recognized, neutrophil infiltration has also been assessed in many clinical studies. In these studies, airway neutrophilia was associated with asthma severity. Importantly, neutrophilia also correlates with asthma that is refractory to corticosteroids, the mainstay of asthma treatment. However, it is now increasingly recognized that neutrophils are a heterogeneous population, and a more precise phenotyping of these cells may help delineate different subtypes of asthma. Here, we review current knowledge of the role of neutrophils in asthma and highlight future avenues of research in this field.
Collapse
Affiliation(s)
- Anuradha Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; University of Pittsburgh Asthma Institute@UPMC/UPSOM, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Jay K Kolls
- Richard King Mellon Institute for Pediatric Research, Children's Hospital of Pittsburgh at University of Pittsburgh Medical Center/University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
40
|
H1N1 influenza virus infection results in adverse pregnancy outcomes by disrupting tissue-specific hormonal regulation. PLoS Pathog 2017; 13:e1006757. [PMID: 29176767 PMCID: PMC5720832 DOI: 10.1371/journal.ppat.1006757] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 12/07/2017] [Accepted: 11/17/2017] [Indexed: 01/06/2023] Open
Abstract
Increased susceptibility to influenza virus infection during pregnancy has been attributed to immunological changes occurring before and during gestation in order to “tolerate” the developing fetus. These systemic changes are most often characterized by a suppression of cell-mediated immunity and elevation of humoral immune responses referred to as the Th1-Th2 shift. However, the underlying mechanisms which increase pregnant mothers’ risk following influenza virus infection have not been fully elucidated. We used pregnant BALB/c mice during mid- to late gestation to determine the impact of a sub-lethal infection with A/Brisbane/59/07 H1N1 seasonal influenza virus on completion of gestation. Maternal and fetal health status was closely monitored and compared to infected non-pregnant mice. Severity of infection during pregnancy was correlated with premature rupture of amniotic membranes (PROM), fetal survival and body weight at birth, lung viral load and degree of systemic and tissue inflammation mediated by innate and adaptive immune responses. Here we report that influenza virus infection resulted in dysregulation of inflammatory responses that led to pre-term labor, impairment of fetal growth, increased fetal mortality and maternal morbidity. We observed significant compartment-specific immune responses correlated with changes in hormonal synthesis and regulation. Dysregulation of progesterone, COX-2, PGE2 and PGF2α expression in infected pregnant mice was accompanied by significant remodeling of placental architecture and upregulation of MMP-9 early after infection. Collectively these findings demonstrate the potential of a seasonal influenza virus to initiate a powerful pro-abortive mechanism with adverse outcomes in fetal health. Maternal immunology is finely balanced to maintain a tolerant and supportive molecular environment for the developing fetus while continuing surveillance against foreign microbial threats. Influenza viral infection during pregnancy is a significant clinical risk for mothers and their newborns, increasing hospitalization, preterm birth, low birth weight, and maternal and neonatal deaths worldwide. In a mouse pregnancy model, we show how influenza virus infection disrupts the delicate and interconnected cytokine and hormonal signaling pathways that respond to respiratory pathogens. The health of mothers and offspring was impacted in our study, after pregnant mothers’ lung and placental architecture was compromised by infection. Influenza virus infection increased the stress on the mother’s body already present due to pregnancy, or reversed the hormonal environment required to establish and maintain healthy pregnancy. By dissecting the effects of inflammation post-infection throughout the mother’s anatomy, we can tailor anti-inflammatory treatments for the pregnant population. Also, thorough knowledge of immune responses will assist in tailoring vaccine design and dosage for this delicate period of women’s immunological and reproductive health.
Collapse
|
41
|
Döring Y, Megens R, Soehnlein O, Drechsler M. Neutrophilic granulocytes – promiscuous accelerators of atherosclerosis. Thromb Haemost 2017; 106:839-48. [DOI: 10.1160/th11-07-0501] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 09/28/2011] [Indexed: 12/23/2022]
Abstract
SummaryNeutrophils, as part of the innate immune system, are classically described to be main actors during the onset of inflammation enforcing rapid neutralisation and clearance of pathogens. Besides their wellstudied role in acute inflammatory processes, recent advances strongly indicate a so far underappreciated importance of neutrophils in initiation and development of atherosclerosis. This review focuses on current findings on the role of neutrophils in atherosclerosis. As pro-inflammatory mechanisms of neutrophils have primarily been studied in the microvascular environment; we here aim at translating these into the context of macrovascular inflammation in atherosclerosis. Since much of the pro-inflammatory activities of neutrophils stem from instructing neighbouring cell types, we highlight the promiscuous interplay between neutrophils and platelets, monocytes, T lymphocytes, and dendritic cells and its possible relevance to atherosclerosis.
Collapse
|
42
|
Lokau J, Agthe M, Flynn CM, Garbers C. Proteolytic control of Interleukin-11 and Interleukin-6 biology. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017. [DOI: 10.1016/j.bbamcr.2017.06.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
43
|
Jingushi K, Uemura M, Ohnishi N, Nakata W, Fujita K, Naito T, Fujii R, Saichi N, Nonomura N, Tsujikawa K, Ueda K. Extracellular vesicles isolated from human renal cell carcinoma tissues disrupt vascular endothelial cell morphology via azurocidin. Int J Cancer 2017; 142:607-617. [DOI: 10.1002/ijc.31080] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 09/18/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Kentaro Jingushi
- Laboratory of Molecular and Cellular Physiology; Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka; Suita Osaka 565-0871 Japan
- Department of Therapeutic Urologic Oncology; Osaka University, Graduate School of Medicine, 2-2 Yamadaoka; Suita 565-0871 Japan
| | - Motohide Uemura
- Department of Therapeutic Urologic Oncology; Osaka University, Graduate School of Medicine, 2-2 Yamadaoka; Suita 565-0871 Japan
- Department of Urology; Osaka University, Graduate School of Medicine, 2-2 Yamadaoka; Suita 565-0871 Japan
| | - Naomi Ohnishi
- Project for Personalized Cancer Medicine, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake; Koto-ku Tokyo 135-8550 Japan
| | - Wataru Nakata
- Department of Urology; Osaka University, Graduate School of Medicine, 2-2 Yamadaoka; Suita 565-0871 Japan
| | - Kazutoshi Fujita
- Department of Urology; Osaka University, Graduate School of Medicine, 2-2 Yamadaoka; Suita 565-0871 Japan
| | - Takuya Naito
- Laboratory of Molecular and Cellular Physiology; Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka; Suita Osaka 565-0871 Japan
| | - Risa Fujii
- Project for Personalized Cancer Medicine, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake; Koto-ku Tokyo 135-8550 Japan
| | - Naomi Saichi
- Project for Personalized Cancer Medicine, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake; Koto-ku Tokyo 135-8550 Japan
| | - Norio Nonomura
- Department of Urology; Osaka University, Graduate School of Medicine, 2-2 Yamadaoka; Suita 565-0871 Japan
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular Physiology; Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka; Suita Osaka 565-0871 Japan
| | - Koji Ueda
- Project for Personalized Cancer Medicine, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake; Koto-ku Tokyo 135-8550 Japan
| |
Collapse
|
44
|
Neutrophils Are Critical for Myelin Removal in a Peripheral Nerve Injury Model of Wallerian Degeneration. J Neurosci 2017; 37:10258-10277. [PMID: 28912156 DOI: 10.1523/jneurosci.2085-17.2017] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/06/2017] [Accepted: 09/09/2017] [Indexed: 12/31/2022] Open
Abstract
Wallerian degeneration (WD) is considered an essential preparatory stage to the process of axonal regeneration. In the peripheral nervous system, infiltrating monocyte-derived macrophages, which use the chemokine receptor CCR2 to gain entry to injured tissues from the bloodstream, are purportedly necessary for efficient WD. However, our laboratory has previously reported that myelin clearance in the injured sciatic nerve proceeds unhindered in the Ccr2-/- mouse model. Here, we extensively characterize WD in male Ccr2-/- mice and identify a compensatory mechanism of WD that is facilitated primarily by neutrophils. In response to the loss of CCR2, injured Ccr2-/- sciatic nerves demonstrate prolonged expression of neutrophil chemokines, a concomitant extended increase in the accumulation of neutrophils in the nerve, and elevated phagocytosis by neutrophils. Neutrophil depletion substantially inhibits myelin clearance after nerve injury in both male WT and Ccr2-/- mice, highlighting a novel role for these cells in peripheral nerve degeneration that spans genotypes.SIGNIFICANCE STATEMENT The accepted view in the basic and clinical neurosciences is that the clearance of axonal and myelin debris after a nerve injury is directed primarily by inflammatory CCR2+ macrophages. However, we demonstrate that this clearance is nearly identical in WT and Ccr2-/- mice, and that neutrophils replace CCR2+ macrophages as the primary phagocytic cell. We find that neutrophils play a major role in myelin clearance not only in Ccr2-/- mice but also in WT mice, highlighting their necessity during nerve degeneration in the peripheral nervous system. These degeneration studies may propel improvements in nerve regeneration and draw critical parallels to mechanisms of nerve degeneration and regeneration in the CNS and in the context of peripheral neuropathies.
Collapse
|
45
|
Caster DJ, Powell DW, Miralda I, Ward RA, McLeish KR. Re-Examining Neutrophil Participation in GN. J Am Soc Nephrol 2017; 28:2275-2289. [PMID: 28620081 DOI: 10.1681/asn.2016121271] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Significant advances in understanding the pathogenesis of GN have occurred in recent decades. Among those advances is the finding that both innate and adaptive immune cells contribute to the development of GN. Neutrophils were recognized as key contributors in early animal models of GN, at a time when the prevailing view considered neutrophils to function as nonspecific effector cells that die quickly after performing antimicrobial functions. However, advances over the past two decades have shown that neutrophil functions are more complex and sophisticated. Specifically, research has revealed that neutrophil survival is regulated by the inflammatory milieu and that neutrophils demonstrate plasticity, mediate microbial killing through previously unrecognized mechanisms, demonstrate transcriptional activity leading to the release of cytokines and chemokines, interact with and regulate cells of the innate and adaptive immune systems, and contribute to the resolution of inflammation. Therefore, neutrophil participation in glomerular diseases deserves re-evaluation. In this review, we describe advances in understanding classic neutrophil functions, review the expanded roles of neutrophils in innate and adaptive immune responses, and summarize current knowledge of neutrophil contributions to GN.
Collapse
Affiliation(s)
- Dawn J Caster
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky, .,Nephrology Section, Medicine Service, Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky, and
| | - David W Powell
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky
| | - Irina Miralda
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Richard A Ward
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky
| | - Kenneth R McLeish
- Division of Nephrology and Hypertension, Department of Medicine, University of Louisville School of Medicine, Louisville, Kentucky.,Nephrology Section, Medicine Service, Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky, and
| |
Collapse
|
46
|
Graubardt N, Vugman M, Mouhadeb O, Caliari G, Pasmanik-Chor M, Reuveni D, Zigmond E, Brazowski E, David E, Chappell-Maor L, Jung S, Varol C. Ly6C hi Monocytes and Their Macrophage Descendants Regulate Neutrophil Function and Clearance in Acetaminophen-Induced Liver Injury. Front Immunol 2017; 8:626. [PMID: 28620383 PMCID: PMC5451509 DOI: 10.3389/fimmu.2017.00626] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/10/2017] [Indexed: 12/13/2022] Open
Abstract
Monocyte-derived macrophages (MoMF) play a pivotal role in the resolution of acetaminophen-induced liver injury (AILI). Timely termination of neutrophil activity and their clearance are essential for liver regeneration following injury. Here, we show that infiltrating Ly6Chi monocytes, their macrophage descendants, and neutrophils spatially and temporally overlap in the centrilobular necrotic areas during the necroinflammatory and resolution phases of AILI. At the necroinflammatory phase, inducible ablation of circulating Ly6Chi monocytes resulted in reduced numbers and fractions of reactive oxygen species (ROS)-producing neutrophils. In alignment with this, neutrophils sorted from monocyte-deficient livers exhibited reduced expression of NADPH oxidase 2. Moreover, human CD14+ monocytes stimulated with lipopolysaccharide or hepatocyte apoptotic bodies directly induced ROS production by cocultured neutrophils. RNA-seq-based transcriptome profiling of neutrophils from Ly6Chi monocyte-deficient versus normal livers revealed 449 genes that were differentially expressed with at least twofold change (p ≤ 0.05). In the absence of Ly6Chi monocytes, neutrophils displayed gene expression alterations associated with decreased innate immune activity and increased cell survival. At the early resolution phase, Ly6Chi monocytes differentiated into ephemeral Ly6Clo MoMF and their absence resulted in significant accumulation of late apoptotic neutrophils. Further gene expression analysis revealed the induced expression of a specific repertoire of bridging molecules and receptors involved with apoptotic cell clearance during the transition from Ly6Chi monocytes to MoMF. Collectively, our findings establish a phase-dependent task division between liver-infiltrating Ly6Chi monocytes and their MoMF descendants with the former regulating innate immune functions and cell survival of neutrophils and the later neutrophil clearance.
Collapse
Affiliation(s)
- Nadine Graubardt
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Milena Vugman
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Odelia Mouhadeb
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Department of Clinical Microbiology and Immunology, The Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Gabriele Caliari
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Metsada Pasmanik-Chor
- Bioinformatics Unit, G. S. Wise Faculty of Life Science, Tel-Aviv University, Tel Aviv, Israel
| | - Debby Reuveni
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Ehud Zigmond
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Eli Brazowski
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Chen Varol
- The Research Center for Digestive Tract and Liver Diseases, Tel-Aviv Sourasky Medical Center and Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Department of Clinical Microbiology and Immunology, The Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
47
|
Mark Welch DB, Jauch A, Langowski J, Olins AL, Olins DE. Transcriptomes reflect the phenotypes of undifferentiated, granulocyte and macrophage forms of HL-60/S4 cells. Nucleus 2017; 8:222-237. [PMID: 28152343 DOI: 10.1080/19491034.2017.1285989] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
To understand the chromatin changes underlying differential gene expression during induced differentiation of human leukemic HL-60/S4 cells, we conducted RNA-Seq analysis on quadruplicate cultures of undifferentiated, granulocytic- and macrophage-differentiated cell forms. More than half of mapped genes exhibited altered transcript levels in the differentiated cell forms. In general, more genes showed increased mRNA levels in the granulocytic form and in the macrophage form, than showed decreased levels. The majority of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were significantly enriched in genes that exhibited differential transcript levels after either RA or TPA treatment. Changes in transcript levels for groups of genes with characteristic protein phenotypes, such as genes encoding cytoplasmic granular proteins, nuclear envelope and cytoskeletal proteins, cell adhesion proteins, and proteins involved in the cell cycle and apoptosis illustrate the profound differences among the various cell states. In addition to the transcriptome analyses, companion karyotyping by M-FISH of undifferentiated HL-60/S4 cells revealed a plethora of chromosome alterations, compared with normal human cells. The present mRNA profiling provides important information related to nuclear shape changes (e.g., granulocyte lobulation), deformability of the nuclear envelope and linkage between the nuclear envelope and cytoskeleton during induced myeloid chromatin differentiation.
Collapse
Affiliation(s)
- David B Mark Welch
- a Josephine Bay Paul Center for Comparative Molecular Biology and Evolution , Marine Biological Laboratory , Woods Hole , MA , USA
| | - Anna Jauch
- b Institute of Human Genetics, University of Heidelberg , Heidelberg , Germany
| | - Jörg Langowski
- c Division Biophysics of Macromolecules, B040 , German Cancer Research Center (DKFZ), TP3 , Heidelberg , Germany
| | - Ada L Olins
- d University of New England, College of Pharmacy , Department of Pharmaceutical Sciences , Portland , ME , USA
| | - Donald E Olins
- d University of New England, College of Pharmacy , Department of Pharmaceutical Sciences , Portland , ME , USA
| |
Collapse
|
48
|
Yang F, Feng C, Zhang X, Lu J, Zhao Y. The Diverse Biological Functions of Neutrophils, Beyond the Defense Against Infections. Inflammation 2017; 40:311-323. [PMID: 27817110 DOI: 10.1007/s10753-016-0458-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Polymorphonuclear neutrophils are among the first defense against infection and closely involved in the initiation of inflammatory response. It is well recognized that this function of neutrophils was mainly mediated by phagocytosis, intracellular degradation, releasing of granules, and formation of neutrophil extracellular traps after sensing dangerous stress. However, accumulating data showed that neutrophils had a variety of important biological functions in both innate and adaptive immunities, far beyond cytotoxicity against pathogens. Neutrophils can differentially switch phenotypes and display distinct subpopulations under different microenvironments. Neutrophils can produce a large variety of cytokines and chemokines upon stimulation. Furthermore, neutrophils directly interact with dendritic cells (DCs), macrophages, natural killer cells, T cells, and B cells so as to either potentiate or down-modulate both innate and adaptive immunity. In the present review, we summarize the recent progress on the functional plasticity and the regulatory ability on immunity of neutrophils in physiological and pathological situations.
Collapse
Affiliation(s)
- Fan Yang
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, Beijing, 100101, China
| | - Chang Feng
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, Beijing, 100101, China
| | - Xiaodong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jun Lu
- Hepatology and Cancer Biotherapy Ward, Beijing YouAn Hospital, Capital Medical University, Beijing, 100069, China.
| | - Yong Zhao
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beichen West Road 1-5, Chaoyang District, Beijing, 100101, China.
| |
Collapse
|
49
|
Fetz AE, Neeli I, Rodriguez IA, Radic MZ, Bowlin GL. Electrospun Template Architecture and Composition Regulate Neutrophil NETosis In Vitro and In Vivo<sup/>. Tissue Eng Part A 2017; 23:1054-1063. [PMID: 28068879 DOI: 10.1089/ten.tea.2016.0452] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Mounting evidence indicates that neutrophils, first responders to an implanted biomaterial, prime the microenvironment for recruited immune cells by secreting factors and releasing neutrophil extracellular traps (NETs) through NETosis. In this study, we investigated the role of electrospun template architecture and composition in regulating NETosis. Electrospun polydioxanone (PDO), collagen type I (COL), and blended PDO-COL templates (PC) were fabricated with small-diameter (0.25-0.35 μm) and large-diameter (1.0-2.00 μm) fibers. Neutrophil-template interactions were evaluated in vitro for 3 and 24 h with human neutrophils, and the PDO templates were studied in vivo (rat subcutaneous model) for 1 and 7 days. Template-bound NETs were quantified by fluorescent microscopy and an On-cell Western assay. The in vitro results indicate that larger fiber diameters reduced NETosis on PDO templates, whereas the incorporation of COL attenuated NETosis independent of fiber diameter. The in vivo results similarly revealed a lower degree of NETs on large-diameter PDO templates at 1 day, resulting in marginal tissue integration of the templates at 7 days. In contrast, the small-diameter PDO templates, which were coated in a large amount of NETs at 24 h in vivo, were surrounded by capsule-like tissue at 7 days. These preliminary in vivo results validate the in vitro model and signify NETosis as a potentially significant physiological response and a critical preconditioning event for the innate immune response to templates. In conclusion, these results demonstrate the importance of characterizing the neutrophil's acute confrontation with biomaterials to engineer templates capable of promoting in situ regeneration.
Collapse
Affiliation(s)
- Allison E Fetz
- 1 Department of Biomedical Engineering, University of Memphis , Memphis, Tennessee
| | - Indira Neeli
- 2 Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Isaac A Rodriguez
- 1 Department of Biomedical Engineering, University of Memphis , Memphis, Tennessee
| | - Marko Z Radic
- 2 Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Gary L Bowlin
- 1 Department of Biomedical Engineering, University of Memphis , Memphis, Tennessee
| |
Collapse
|
50
|
Druhan LJ, Lance A, Li S, Price AE, Emerson JT, Baxter SA, Gerber JM, Avalos BR. Leucine Rich α-2 Glycoprotein: A Novel Neutrophil Granule Protein and Modulator of Myelopoiesis. PLoS One 2017; 12:e0170261. [PMID: 28081565 PMCID: PMC5233425 DOI: 10.1371/journal.pone.0170261] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 12/30/2016] [Indexed: 02/01/2023] Open
Abstract
Leucine-rich α2 glycoprotein (LRG1), a serum protein produced by hepatocytes, has been implicated in angiogenesis and tumor promotion. Our laboratory previously reported the expression of LRG1 in murine myeloid cell lines undergoing neutrophilic granulocyte differentiation. However, the presence of LRG1 in primary human neutrophils and a role for LRG1 in regulation of hematopoiesis have not been previously described. Here we show that LRG1 is packaged into the granule compartment of human neutrophils and secreted upon neutrophil activation to modulate the microenvironment. Using immunofluorescence microscopy and direct biochemical measurements, we demonstrate that LRG1 is present in the peroxidase-negative granules of human neutrophils. Exocytosis assays indicate that LRG1 is differentially glycosylated in neutrophils, and co-released with the secondary granule protein lactoferrin. Like LRG1 purified from human serum, LRG1 secreted from activated neutrophils also binds cytochrome c. We also show that LRG1 antagonizes the inhibitory effects of TGFβ1 on colony growth of human CD34+ cells and myeloid progenitors. Collectively, these data invoke an additional role for neutrophils in innate immunity that has not previously been reported, and suggest a novel mechanism whereby neutrophils may modulate the microenvironment via extracellular release of LRG1.
Collapse
Affiliation(s)
- Lawrence J. Druhan
- The Department of Hematologic Oncology, The Levine Cancer Institute, Carolinas HealthCare System, Charlotte, North Carolina, United States of America
| | - Amanda Lance
- The Department of Hematologic Oncology, The Levine Cancer Institute, Carolinas HealthCare System, Charlotte, North Carolina, United States of America
| | - Shimena Li
- The University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Andrea E. Price
- The Department of Hematologic Oncology, The Levine Cancer Institute, Carolinas HealthCare System, Charlotte, North Carolina, United States of America
| | - Jacob T. Emerson
- The Department of Hematologic Oncology, The Levine Cancer Institute, Carolinas HealthCare System, Charlotte, North Carolina, United States of America
| | - Sarah A. Baxter
- The Department of Hematologic Oncology, The Levine Cancer Institute, Carolinas HealthCare System, Charlotte, North Carolina, United States of America
| | - Jonathan M. Gerber
- The Department of Hematologic Oncology, The Levine Cancer Institute, Carolinas HealthCare System, Charlotte, North Carolina, United States of America
| | - Belinda R. Avalos
- The Department of Hematologic Oncology, The Levine Cancer Institute, Carolinas HealthCare System, Charlotte, North Carolina, United States of America
- * E-mail:
| |
Collapse
|