1
|
Pednekar K, Minnee J, de Vries IJM, Prakash J. Targeted nanomedicine for reprogramming the tumor innate immune system: From bench to bedside. Eur J Pharm Biopharm 2024; 204:114510. [PMID: 39307440 DOI: 10.1016/j.ejpb.2024.114510] [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: 07/02/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/27/2024]
Abstract
Tumor-associated innate immune cells such as tumor-associated macrophages, neutrophils, dendritic cells play a crucial role in tumor progression, angiogenesis and metastasis. These cells also control the efficacy of chemotherapy and immunotherapy by inducing drug resistance and immunosuppression, leading to therapeutic failures. Therefore, targeting the tumor-associated innate immune cells has gained high attention for the development of effective cancer therapy. Nanomedicine based strategies to target these cells are highly relevant and can be used to reprogram these cells. In this review, we discuss the fundamental roles of the tumor-associated innate immune cells in the tumor microenvironment and different strategies to modulate them. Then, nanomedicine-based strategies to target different tumor innate immune cells are explained in detail. While the clinical development of the targeted nanomedicine remains a great challenge in practice, we have provided our perspectives on various factors such as pharmaceutical aspects, preclinical testing and biological aspects which are crucial to consider before translating these targeting strategies to clinics.
Collapse
Affiliation(s)
- Kunal Pednekar
- Engineered Therapeutics, Department of Advanced Organ bioengineering and Therapeutics, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Julia Minnee
- Department of Medical BioSciences (MBS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Medical BioSciences (MBS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jai Prakash
- Engineered Therapeutics, Department of Advanced Organ bioengineering and Therapeutics, Technical Medical Centre, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
2
|
Amsler J, Everts-Graber J, Martin KR, Roccabianca A, Lopes C, Tourneur L, Mocek J, Karras A, Naccache JM, Bonnotte B, Samson M, Hanslik T, Puéchal X, Terrier B, Guillevin L, Néel A, Mouthon L, Witko-Sarsat V. Dysregulation of neutrophil oxidant production and interleukin-1-related cytokines in granulomatosis with polyangiitis. Rheumatology (Oxford) 2024; 63:2249-2258. [PMID: 37947315 DOI: 10.1093/rheumatology/kead578] [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: 06/18/2023] [Revised: 09/28/2023] [Accepted: 10/06/2023] [Indexed: 11/12/2023] Open
Abstract
OBJECTIVES Neutrophils play a key role in ANCA-associated vasculitis, both as targets of autoimmunity and as facilitators of vascular damage. In granulomatosis with polyangiitis (GPA), the data regarding the production of reactive oxygen species (ROS) in neutrophils are unclear. Further, recent data suggests that ROS production could have an anti-inflammatory effect through the regulation of inflammasomes and IL-1-related cytokines. We aimed to analyse ROS production in neutrophils from patients with GPA and investigate its association with IL-1-related cytokines and the autoantigen PR3. METHODS Seventy-two GPA patients with disease flare were included in the NEUTROVASC prospective cohort study. ROS production in whole blood of patients with active GPA was evaluated and compared with that in the same patients in remission or healthy controls. Associations between ROS production, PR3 membrane expression on neutrophils, serum levels of IL-1-related cytokines as well as inflammasome-related proteins were analysed. RESULTS We observed a robust defect in ROS production by neutrophils from patients with active GPA compared with healthy controls, independent of glucocorticoid treatment. Serum levels of IL-1-related cytokines were significantly increased in GPA patients, particularly in patients with kidney involvement, and levels of these cytokines returned to normal after patients achieved remission. Further, inflammasome-related proteins were significantly dysregulated in the cytosol of neutrophils as well as the serum from GPA patients. CONCLUSION Our data suggests that ROS production and regulation of inflammasomes in neutrophils from patients with GPA are disturbed and may be a potential therapeutic target. TRIAL REGISTRATION ClinicalTrials.gov, https://www.clinicaltrials.gov, NCT01862068.
Collapse
Affiliation(s)
- Jennifer Amsler
- INSERM U1016, Institut Cochin, CNRS UMR 8104, Paris, France
- Université Paris Cité, Paris, France
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Judith Everts-Graber
- INSERM U1016, Institut Cochin, CNRS UMR 8104, Paris, France
- Université Paris Cité, Paris, France
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Katherine R Martin
- INSERM U1016, Institut Cochin, CNRS UMR 8104, Paris, France
- Université Paris Cité, Paris, France
- Inflammation Division, WEHI, and Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Arnaud Roccabianca
- INSERM U1016, Institut Cochin, CNRS UMR 8104, Paris, France
- Université Paris Cité, Paris, France
| | - Chloé Lopes
- INSERM U1016, Institut Cochin, CNRS UMR 8104, Paris, France
- Université Paris Cité, Paris, France
| | - Léa Tourneur
- INSERM U1016, Institut Cochin, CNRS UMR 8104, Paris, France
- Université Paris Cité, Paris, France
| | - Julie Mocek
- INSERM U1016, Institut Cochin, CNRS UMR 8104, Paris, France
- Université Paris Cité, Paris, France
| | - Alexandre Karras
- Department of Nephrology, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Jean-Marc Naccache
- Department of Pulmonology-Allergology-Thoracic Oncology, Groupe Hospitalier Paris Saint Joseph, Paris, France
| | - Bernard Bonnotte
- Service de Médecine Interne et Immunologie Clinique, CHU Dijon, Dijon, France
| | - Maxime Samson
- Service de Médecine Interne et Immunologie Clinique, CHU Dijon, Dijon, France
| | - Thomas Hanslik
- Service de Médecine Interne, Hôpital Ambroise-Paré, AP-HP, Boulogne Billancourt, France
| | - Xavier Puéchal
- INSERM U1016, Institut Cochin, CNRS UMR 8104, Paris, France
- Université Paris Cité, Paris, France
- Department of Internal Medicine, Centre de Référence pour les Maladies Systémiques Autoimmunes Rares d'Ile de France, Cochin Hospital, AP-HP, Paris, France
| | - Benjamin Terrier
- INSERM U1016, Institut Cochin, CNRS UMR 8104, Paris, France
- Université Paris Cité, Paris, France
- Department of Internal Medicine, Centre de Référence pour les Maladies Systémiques Autoimmunes Rares d'Ile de France, Cochin Hospital, AP-HP, Paris, France
| | - Loïc Guillevin
- INSERM U1016, Institut Cochin, CNRS UMR 8104, Paris, France
- Université Paris Cité, Paris, France
- Department of Internal Medicine, Centre de Référence pour les Maladies Systémiques Autoimmunes Rares d'Ile de France, Cochin Hospital, AP-HP, Paris, France
| | - Antoine Néel
- Service de Médecine Interne, CHU Nantes, Nantes, France
- CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes Université, Nantes, France
| | - Luc Mouthon
- INSERM U1016, Institut Cochin, CNRS UMR 8104, Paris, France
- Université Paris Cité, Paris, France
- Department of Internal Medicine, Centre de Référence pour les Maladies Systémiques Autoimmunes Rares d'Ile de France, Cochin Hospital, AP-HP, Paris, France
| | - Véronique Witko-Sarsat
- INSERM U1016, Institut Cochin, CNRS UMR 8104, Paris, France
- Université Paris Cité, Paris, France
| |
Collapse
|
3
|
Traum A, Jehle S, Waxmann Y, Litmeyer AS, Berghöfer H, Bein G, Dammann R, Perniss A, Burg-Roderfeld M, Sachs UJ, Bayat B. The CD177 c.1291A Allele Leads to a Loss of Membrane Expression and Mimics a CD177-Null Phenotype. Int J Mol Sci 2024; 25:2877. [PMID: 38474126 DOI: 10.3390/ijms25052877] [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: 07/14/2023] [Revised: 02/18/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
CD177 is a glycosyl phosphatidyl inositol (GPI)-linked, neutrophil-specific glycoprotein that in 3-5% of normal individuals is absent from all neutrophils. The molecular mechanism behind the absence of CD177 has not been unravelled completely. Here, we analyse the impact of the recently described CD177 c.1291G>A variant on CD177 expression. Recombinant CD177 c.1291G>A was expressed in HEK293F cells and its expression on the cell surface, inside the cell, and in the culture supernatant was investigated. The CD177 c.1291G>A protein was characterised serologically and its interaction with proteinase 3 (PR3) was demonstrated by confocal laser scanning microscopy. Our experiments show that CD177 c.1291G>A does not interfere with CD177 protein biosynthesis but affects the membrane expression of CD177, leading to very low copy numbers of the protein on the cellular surface. The mutation does not interfere with the ability of the protein to bind PR3 or human polyclonal antibodies against wild-type CD177. Carriers of the c.1291G>A allele are supposed to be phenotyped as CD177-negative, but the protein is present in soluble form. The presence of CD177 c.1291A leads to the production of an unstable CD177 protein and an apparent "CD177-null" phenotype.
Collapse
Affiliation(s)
- Annalena Traum
- Institute for Clinical Immunology, Transfusion Medicine and Haemostasis, Medical Faculty, Justus-Liebig-University, 35390 Giessen, Germany
| | - Stefanie Jehle
- Institute for Clinical Immunology, Transfusion Medicine and Haemostasis, Medical Faculty, Justus-Liebig-University, 35390 Giessen, Germany
| | - Yannick Waxmann
- Institute for Clinical Immunology, Transfusion Medicine and Haemostasis, Medical Faculty, Justus-Liebig-University, 35390 Giessen, Germany
| | - Anne-Sophie Litmeyer
- Institute for Clinical Immunology, Transfusion Medicine and Haemostasis, Medical Faculty, Justus-Liebig-University, 35390 Giessen, Germany
| | - Heike Berghöfer
- Institute for Clinical Immunology, Transfusion Medicine and Haemostasis, Medical Faculty, Justus-Liebig-University, 35390 Giessen, Germany
| | - Gregor Bein
- Institute for Clinical Immunology, Transfusion Medicine and Haemostasis, Medical Faculty, Justus-Liebig-University, 35390 Giessen, Germany
| | - Reinhard Dammann
- Institute for Genetics, Faculty of Biology and Chemistry, Justus-Liebig-University, 35390 Giessen, Germany
| | - Alexander Perniss
- Institute for Anatomy and Cell Biology, German Center for Lung Research, Excellence Cluster Cardio-Pulmonary Institute (CPI), Justus-Liebig-University, 35392 Giessen, Germany
| | - Monika Burg-Roderfeld
- Faculty of Biology and Chemistry, Fresenius University of Applied Sciences, 65510 Idstein, Germany
| | - Ulrich J Sachs
- Institute for Clinical Immunology, Transfusion Medicine and Haemostasis, Medical Faculty, Justus-Liebig-University, 35390 Giessen, Germany
- Department of Thrombosis and Haemostasis, Giessen University Hospital, 35390 Giessen, Germany
| | - Behnaz Bayat
- Institute for Clinical Immunology, Transfusion Medicine and Haemostasis, Medical Faculty, Justus-Liebig-University, 35390 Giessen, Germany
| |
Collapse
|
4
|
Huang F, Lv Y, Liu S, Wu H, Liu Q. Animal models for anti-neutrophil cytoplasmic antibody-associated vasculitis: Are current models good enough? Animal Model Exp Med 2023; 6:452-463. [PMID: 37614099 PMCID: PMC10614129 DOI: 10.1002/ame2.12345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/22/2023] [Indexed: 08/25/2023] Open
Abstract
Antineutrophil cytoplasmic autoantibody (ANCA)-associated vasculitis (AAV) is a rare and severe systemic autoimmune disease characterized by pauci-immune necrotizing inflammation of small blood vessels. AAV involves multiple organ systems throughout the body. Our knowledge of the pathogenesis of AAV has increased considerably in recent years, involving cellular, molecular and genetic factors. Because of the controlled environment with no other confounding factors, animal models are beneficial for studying the mechanistic details of disease development and for providing novel therapeutic targets with fewer toxic side effects. However, the complexity and heterogeneity of AAV make it very difficult to establish a single animal model that can fully represent the entire clinical spectrum found in patients. The aim of this review is to overview the current status of animal models for AAV, outline the pros and cons of methods, and propose potential directions for future research.
Collapse
Affiliation(s)
- Fei Huang
- Department of GeriatricsTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
- Department of General MedicineTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
| | - Yongman Lv
- Department of NephrologyTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
- Department of health management centerTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
| | - Siyang Liu
- Department of NephrologyTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
| | - Hao Wu
- Interdisciplinary Sciences InstituteHuazhong Agricultural UniversityWuhanChina
- State Key Laboratory of Agricultural Microbiology, College of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Qingquan Liu
- Department of NephrologyTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
5
|
Huber A, Jose S, Kassam A, Weghorn KN, Powers-Fletcher M, Sharma D, Mukherjee A, Mathew A, Kulkarni N, Chandramouli S, Alder MN, Madan R. Olfactomedin-4 + neutrophils exacerbate intestinal epithelial damage and worsen host survival after Clostridioides difficile infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.21.553751. [PMID: 37662327 PMCID: PMC10473617 DOI: 10.1101/2023.08.21.553751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Neutrophils are key first responders to Clostridioides difficile infection (CDI). Excessive tissue and blood neutrophils are associated with worse histopathology and adverse outcomes, however their functional role during CDI remains poorly defined. Utilizing intestinal epithelial cell (IEC)-neutrophil co-cultures and a pre-clinical animal model of CDI, we show that neutrophils exacerbate C. difficile -induced IEC injury. We utilized cutting-edge single-cell transcriptomics to illuminate neutrophil subtypes and biological pathways that could exacerbate CDI-associated IEC damage. As such, we have established the first transcriptomics atlas of bone marrow (BM), blood, and colonic neutrophils after CDI. We found that CDI altered the developmental trajectory of BM and blood neutrophils towards populations that exhibit gene signatures associated with pro-inflammatory responses and neutrophil-mediated tissue damage. Similarly, the transcriptomic signature of colonic neutrophils was consistent with hyper-inflammatory and highly differentiated cells that had amplified expression of cytokine-mediated signaling and degranulation priming genes. One of the top 10 variable features in colonic neutrophils was the gene for neutrophil glycoprotein, Olfactomedin 4 (OLFM4). CDI enhanced OLFM4 mRNA and protein expression in neutrophils, and OLFM4 + cells aggregated to areas of severe IEC damage. Compared to uninfected controls, both humans and mice with CDI had higher concentrations of circulating OLFM4; and in mice, OLFM4 deficiency resulted in faster recovery and better survival after infection. Collectively, these studies provide novel insights into neutrophil-mediated pathology after CDI and highlight the pathogenic role of OLFM4 + neutrophils in regulating CDI-induced IEC damage. One Sentence Summary Utilizing single-cell transcriptomics, IEC-epithelial co-cultures, and pre-clinical models of CDI, we have identified a subset of neutrophils that are marked by OLFM4 expression as pathogenic determinants of IEC barrier damage after CDI.
Collapse
|
6
|
Bruserud Ø, Mosevoll KA, Bruserud Ø, Reikvam H, Wendelbo Ø. The Regulation of Neutrophil Migration in Patients with Sepsis: The Complexity of the Molecular Mechanisms and Their Modulation in Sepsis and the Heterogeneity of Sepsis Patients. Cells 2023; 12:cells12071003. [PMID: 37048076 PMCID: PMC10093057 DOI: 10.3390/cells12071003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Common causes include gram-negative and gram-positive bacteria as well as fungi. Neutrophils are among the first cells to arrive at an infection site where they function as important effector cells of the innate immune system and as regulators of the host immune response. The regulation of neutrophil migration is therefore important both for the infection-directed host response and for the development of organ dysfunctions in sepsis. Downregulation of CXCR4/CXCL12 stimulates neutrophil migration from the bone marrow. This is followed by transmigration/extravasation across the endothelial cell barrier at the infection site; this process is directed by adhesion molecules and various chemotactic gradients created by chemotactic cytokines, lipid mediators, bacterial peptides, and peptides from damaged cells. These mechanisms of neutrophil migration are modulated by sepsis, leading to reduced neutrophil migration and even reversed migration that contributes to distant organ failure. The sepsis-induced modulation seems to differ between neutrophil subsets. Furthermore, sepsis patients should be regarded as heterogeneous because neutrophil migration will possibly be further modulated by the infecting microorganisms, antimicrobial treatment, patient age/frailty/sex, other diseases (e.g., hematological malignancies and stem cell transplantation), and the metabolic status. The present review describes molecular mechanisms involved in the regulation of neutrophil migration; how these mechanisms are altered during sepsis; and how bacteria/fungi, antimicrobial treatment, and aging/frailty/comorbidity influence the regulation of neutrophil migration.
Collapse
Affiliation(s)
- Øystein Bruserud
- Leukemia Research Group, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Correspondence:
| | - Knut Anders Mosevoll
- Section for Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Section for Infectious Diseases, Department of Clinical Research, University of Bergen, 5021 Bergen, Norway
| | - Øyvind Bruserud
- Department for Anesthesiology and Intensive Care, Haukeland University Hospital, 5021 Bergen, Norway
| | - Håkon Reikvam
- Leukemia Research Group, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Øystein Wendelbo
- Section for Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Faculty of Health, VID Specialized University, Ulriksdal 10, 5009 Bergen, Norway
| |
Collapse
|
7
|
Matsumoto K, Suzuki K, Yasuoka H, Hirahashi J, Yoshida H, Magi M, Noguchi-Sasaki M, Kaneko Y, Takeuchi T. Longitudinal monitoring of circulating immune cell phenotypes in anti-neutrophil cytoplasmic antibody-associated vasculitis. Autoimmun Rev 2023; 22:103271. [PMID: 36627064 DOI: 10.1016/j.autrev.2023.103271] [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: 12/10/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023]
Abstract
Anti-neutrophil cytoplasmic antibody-associated vasculitis (AAV) is a necrotizing multiorgan autoimmune disease that affects small- to medium-sized blood vessels. Despite the improvements in treatments, half of the patients with AAV still experience disease relapses. In this review, we focus on peripheral leukocyte properties and phenotypes in patients with AAV. In particular, we explore longitudinal changes in circulating immune cell phenotypes during the active phase of the disease and treatment. The numbers and phenotypes of leukocytes in peripheral blood were differs between AAV and healthy controls, AAV in active versus inactive phase, AAV in treatment responders versus non-responders, and AAV with and without severe infection. Therefore, biomarkers detected in peripheral blood immune cells may be useful for longitudinal monitoring of disease activity in AAV.
Collapse
Affiliation(s)
- Kotaro Matsumoto
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Katsuya Suzuki
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hidekata Yasuoka
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; Division of Rheumatology, Department of Internal Medicine, Fujita Health University School of Medicine, Aichi, Japan
| | - Junichi Hirahashi
- Center for General Medicine Education, Keio University School of Medicine, Tokyo, Japan
| | | | - Mayu Magi
- Chugai Pharmaceutical Co. Ltd., Kanagawa, Japan
| | | | - Yuko Kaneko
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Aymonnier K, Amsler J, Lamprecht P, Salama A, Witko‐Sarsat V. The neutrophil: A key resourceful agent in immune‐mediated vasculitis. Immunol Rev 2022; 314:326-356. [PMID: 36408947 DOI: 10.1111/imr.13170] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The term "vasculitis" refers to a group of rare immune-mediated diseases characterized by the dysregulated immune system attacking blood vessels located in any organ of the body, including the skin, lungs, and kidneys. Vasculitides are classified according to the size of the vessel that is affected. Although this observation is not specific to small-, medium-, or large-vessel vasculitides, patients show a high circulating neutrophil-to-lymphocyte ratio, suggesting the direct or indirect involvement of neutrophils in these diseases. As first responders to infection or inflammation, neutrophils release cytotoxic mediators, including reactive oxygen species, proteases, and neutrophil extracellular traps. If not controlled, this dangerous arsenal can injure the vascular system, which acts as the main transport route for neutrophils, thereby amplifying the initial inflammatory stimulus and the recruitment of immune cells. This review highlights the ability of neutrophils to "set the tone" for immune cells and other cells in the vessel wall. Considering both their long-established and newly described roles, we extend their functions far beyond their direct host-damaging potential. We also review the roles of neutrophils in various types of primary vasculitis, including immune complex vasculitis, anti-neutrophil cytoplasmic antibody-associated vasculitis, polyarteritis nodosa, Kawasaki disease, giant cell arteritis, Takayasu arteritis, and Behçet's disease.
Collapse
Affiliation(s)
- Karen Aymonnier
- INSERM U1016, Institut Cochin, Université Paris Cité, CNRS 8104 Paris France
| | - Jennifer Amsler
- INSERM U1016, Institut Cochin, Université Paris Cité, CNRS 8104 Paris France
| | - Peter Lamprecht
- Department of Rheumatology and Clinical Immunology University of Lübeck Lübeck Germany
| | - Alan Salama
- Department of Renal Medicine, Royal Free Hospital University College London London UK
| | | |
Collapse
|
9
|
Jurgec S, Jezernik G, Gorenjak M, Büdefeld T, Potočnik U. Meta-Analytic Comparison of Global RNA Transcriptomes of Acute and Chronic Myeloid Leukemia Cells Reveals Novel Gene Candidates Governing Myeloid Malignancies. Cancers (Basel) 2022; 14:cancers14194681. [PMID: 36230605 PMCID: PMC9562668 DOI: 10.3390/cancers14194681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Despite advances in the understanding of genetic risk factors and molecular mechanisms underlying acute myeloid leukemia (AML) and chronic myeloid leukemia (CML), clinical outcomes of current therapies in terms of disease relapse and mortality rate pose a great economic and social burden. To overcome this, the identification of new molecular prognostic biomarkers and pharmacological targets is crucial. Recent studies have suggested that AML and CML may share common pathogenic mechanisms and cellular substrates. To this end, in the present study, global transcriptome profiles of AML and CML at the molecular and cellular level were directly compared using a combination of meta-analysis and modern statistics, and novel candidate genes and specific biological processes associated with the pathogenesis of AML and CML were characterized. Our study significantly improves our current understanding of myeloid leukemia and will help develop new therapeutic targets and biomarkers for disease progression, management and treatment response. Abstract Background: Acute myeloid leukemia (AML) and chronic myeloid leukemia (CML) represent a group of hematological malignancies characterized by the pathogenic clonal expansion of leukemic myeloid cells. The diagnosis and clinical outcome of AML and CML are complicated by genetic heterogeneity of disease; therefore, the identification of novel molecular biomarkers and pharmacological targets is of paramount importance. Methods: RNA-seq-based transcriptome data from a total of five studies were extracted from NCBI GEO repository and subjected to an in-depth bioinformatics analysis to identify differentially expressed genes (DEGs) between AML and CML. A systemic literature survey and functional gene ontology (GO) enrichment analysis were performed for the top 100 DEGs to identify novel candidate genes and biological processes associated with AML and CML. Results: LINC01554, PTMAP12, LOC644936, RPS27AP20 and FAM133CP were identified as novel risk genes for AML and CML. GO enrichment analysis showed that DEGs were significantly associated with pre-RNA splicing, reactive oxygen species and glycoprotein metabolism, the cellular endomembrane system, neutrophil migration and antimicrobial immune response. Conclusions: Our study revealed novel biomarkers and specific biological processes associated with AML and CML. Further studies are required to evaluate their value as molecular targets for managing and treating the myeloid malignancies.
Collapse
Affiliation(s)
- Staša Jurgec
- Center for Human Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
- Laboratory for Biochemistry, Molecular Biology and Genomics, Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
| | - Gregor Jezernik
- Center for Human Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Mario Gorenjak
- Center for Human Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Tomaž Büdefeld
- Center for Human Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Uroš Potočnik
- Center for Human Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
- Laboratory for Biochemistry, Molecular Biology and Genomics, Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
- Department for Science and Research, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia
- Correspondence: ; Tel.: +386-2-2345-854
| |
Collapse
|
10
|
Shiezadeh F, Azami N, Arab HR, Rezaee SAR, Moeintaghavi A, Banihashemrad A. Evaluation of Neutrophilic Receptors; CXCL8 and CXCR2 in Patients with Chronic Periodontitis Compared to Healthy Subjects by Real Time PCR Method. Open Dent J 2022. [DOI: 10.2174/18742106-v16-e2202241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Objective:
This study aimed to evaluate the levels of CXCL8 and its receptor (CXCR2) in gingival tissue neutrophils from patients with chronic periodontitis compared to periodontal healthy subjects using Real Time PCR method.
Methods:
The test group consisted of 21 patients with chronic periodontitis and the control group consisted of 18 healthy individuals. In both groups, gingival tissue samples were obtained during periodontal surgery.CXCL8 and CXCR2 RNA in tissue samples were examined by PCR method, and then the levels of genes expression were measured. Mann-Whitney U nonparametric test was used for statistical analysis.
Results:
CXCL8 gene expression in the gingival tissue of the test group with chronic periodontitis was significantly higher than the control group (p=0.028). CXCR2 gene expression in the gingival tissue of the test group with chronic periodontitis was significantly lower than the control group (p=0.043). In both test and control groups, there was a negative correlation between CXCL8 and CXCR2 gene expression. This correlation was statistically significant in the test group (p=0.001), but there was no significant correlation in the control group (p=0.431).
Conclusion:
The results of this present study suggested that the level of gene expression for CXCL8 was greater in patients with chronic periodontitis and CXCR2 was greater in healthy individuals. Although in people with chronic periodontitis, CXCR2 decreases slightly as CXCL8 levels increase.
Collapse
|
11
|
Competitively disrupting the neutrophil-specific receptor-autoantigen CD177:proteinase 3 membrane complex reduces anti-PR3 antibody-induced neutrophil activation. J Biol Chem 2022; 298:101598. [PMID: 35063507 PMCID: PMC8857647 DOI: 10.1016/j.jbc.2022.101598] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 01/15/2023] Open
Abstract
CD177 is a neutrophil-specific receptor presenting the proteinase 3 (PR3) autoantigen on the neutrophil surface. CD177 expression is restricted to a neutrophil subset, resulting in CD177pos/mPR3high and CD177neg/mPR3low populations. The CD177pos/mPR3high subset has implications for antineutrophil cytoplasmic autoantibody (ANCA)-associated autoimmune vasculitis, wherein patients harbor PR3-specific ANCAs that activate neutrophils for degranulation. Here, we generated high-affinity anti-CD177 monoclonal antibodies, some of which interfered with PR3 binding to CD177 (PR3 "blockers") as determined by surface plasmon resonance spectroscopy and used them to test the effect of competing PR3 from the surface of CD177pos neutrophils. Because intact anti-CD177 antibodies also caused neutrophil activation, we prepared nonactivating Fab fragments of a PR3 blocker and nonblocker that bound specifically to CD177pos neutrophils. We observed that Fab blocker clone 40, but not nonblocker clone 80, dose-dependently reduced anti-PR3 antibody binding to CD177pos neutrophils. Importantly, preincubation with clone 40 significantly reduced respiratory burst in primed neutrophils challenged with either monoclonal antibodies to PR3 or PR3-ANCA immunoglobulin G from ANCA-associated autoimmune vasculitis patients. After separating the two CD177/mPR3 neutrophil subsets from individual donors by magnetic sorting, we found that PR3-ANCAs provoked significantly more superoxide production in CD177pos/mPR3high than in CD177neg/mPR3low neutrophils, and that anti-CD177 Fab clone 40 reduced the superoxide production of CD177pos cells to the level of the CD177neg cells. Our data demonstrate the importance of the CD177:PR3 membrane complex in maintaining a high ANCA epitope density and thereby underscore the contribution of CD177 to the severity of PR3-ANCA diseases.
Collapse
|
12
|
Goldmann WH. Proteinase 3 associated with Wegener's Granulomatosis. Cell Biol Int 2021; 46:548-553. [PMID: 34957648 DOI: 10.1002/cbin.11757] [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: 10/31/2021] [Accepted: 12/22/2021] [Indexed: 11/07/2022]
Abstract
Wegener's granulomatosis (WG) is a form of systemic vasculitis characterized by granulomatous inflammation of the upper and lower airways, vasculitis, and necrotizing glomerulonephritis. It is strongly associated with anti-neutrophil cytoplasmic antibodies against proteinase 3 (PR3-ANCAs). Various in vitro observations provided strong evidence that autoimmune PR3-ANCAs are directly involved in glomerular and vascular inflammation. However, little is known about the pathogenic significance of PR3-ANCAs in vivo. Therefore, the generation of animal models helped to validate the suggested autoimmune origin and pathophysiology in WG. To characterize and improve the models, numerous studies were carried out to elucidate the effect of mouse/rat PR3-ANCAs on neutrophil function as well as the role of CD4/CD8 in T and B cells and antibodies in the pathogenesis of the disease. Understanding the pathogenesis is therefore critical to relate these models to human studies hoping that they will be useful for better insight of Wegener's granulomatosis and the development of specific therapies for the disease. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Wolfgang H Goldmann
- Department of Biophysics, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
13
|
Lee NK, Kim SN, Park CG. Immune cell targeting nanoparticles: a review. Biomater Res 2021; 25:44. [PMID: 34930494 PMCID: PMC8690904 DOI: 10.1186/s40824-021-00246-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/28/2021] [Indexed: 02/07/2023] Open
Abstract
Immune cells are attractive targets for therapy as they are direct participants in a variety of diseases. Delivering a therapeutic agent only to cells that act on a disease by distinguishing them from other cells has the advantage of concentrating the therapeutic effect and lowering systemic side effects. Distinguishing each immune cell from other immune cells to deliver substances, including drugs and genes, can be achieved using nanotechnology. And also nanoparticles can ensure in vivo stability and sustained drug release. In addition, there is an ease of surface modification, which is an important characteristic that can be utilized in targeted drug delivery systems. This characteristic allows us to utilize various properties that are specifically expressed in each immune cell. A number of studies have delivered various substances specifically to immune cells through surface engineering with active target ligands that can target each immune cell and enzyme-responsive coating, and demonstrated high therapeutic effects compared to conventional treatments. Progress in research on target delivery has been suggested to be a breakthrough for the treatments of various diseases, including cancer treatment.
Collapse
Affiliation(s)
- Na Kyeong Lee
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Se-Na Kim
- Institute of Medical & Biological Engineering, Medical Research Center, Seoul National University, Seoul, 03080, Republic of Korea
| | - Chun Gwon Park
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea.
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi, 16419, Republic of Korea.
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea.
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, Gyeonggi, 16419, Republic of Korea.
| |
Collapse
|
14
|
The Regulation of Neutrophil Extracellular Trap-induced Tissue Damage by Human CD177. Transplant Direct 2021; 7:e734. [PMID: 34549086 PMCID: PMC8439991 DOI: 10.1097/txd.0000000000001175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/30/2021] [Accepted: 04/09/2021] [Indexed: 01/06/2023] Open
Abstract
Background Neutrophil-induced tissue damage contributes to the rejection in xenotransplantation. Therefore, suppressing neutrophil function could be effective in suppressing xenogeneic rejection. In a previous study, we demonstrated that the ectopic expression of human cluster of differentiation 31 (CD31) on porcine endothelial cells (PEC) significantly suppressed neutrophil-mediated cytotoxicity through the homophilic binding of CD31. Cluster of differentiation 177 (CD177) was recently reported to be a high-affinity heterophilic binding partner for CD31 on endothelial cells. Thus, we hypothesized that human CD177 on PEC might induce a stronger suppression in neutrophil-mediated cytotoxicity compared with CD31. In this study, the inhibitory function of human CD177 on PEC in neutrophil-mediated cytotoxicity was investigated. Methods PEC were transfected with a cloning plasmid containing cDNA inserts that encoded for hCD177 and hCD31 genes. Neutrophil-induced cytotoxicity was evaluated by flow cytometry after coculturing with PEC or PEC/CD177 in the presence of phorbol 12-myristate 13-acetate. To elucidate the mechanisms responsible for hCD177-induced suppression, the phosphorylation of src homology region 2 domain containing phosphatase 1 was measured by immunoblot analysis. Results Human CD177 on PEC induced a significant reduction in neutrophil-induced cytotoxicity. In addition, CD177 on PEC induced a significant increase in the phosphorylation of src homology region 2 domain-containing phosphatase 1 in neutrophils and suppressed NETosis. Conclusions These findings suggest that human CD177 suppresses neutrophil-mediated cytotoxicity through the inhibition of NETosis.
Collapse
|
15
|
Hui W, Zhang W, Liu C, Wan S, Sun W, Su L. Alterations of Signaling Pathways in Essential Thrombocythemia with Calreticulin Mutation. Cancer Manag Res 2021; 13:6231-6238. [PMID: 34393515 PMCID: PMC8357313 DOI: 10.2147/cmar.s316919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/26/2021] [Indexed: 12/25/2022] Open
Abstract
Purpose Though mutations of the calreticulin (CALR) gene have been identified in essential thrombocythemia patients, the detailed mechanisms for CALR mutations have not been completely clarified. Our study is aimed at characterizing alteration of protein expression in ET patients with mutated CALRdel52 and further recognizing possible involvement of signaling pathways associated with CALR mutations. Patients and Methods Protein pathway array was performed to analyze the expression levels of proteins involved in various signaling pathways in peripheral blood neutrophils from 18 ET patients with mutated CALRdel52, 20 ET patients with JAK2V617F mutation and 20 controls. Results We found 20 proteins differentially expressed in ET patients with mutated CALRdel52 compared with healthy controls. These proteins were associated with molecular mechanisms of cancer in ingenuity pathways analysis (IPA) network. We identified top ten canonical pathways which including apoptotic pathways and cellular cytokine pathways might participate in pathogenesis of ET with mutated CALRdel52. Additionally, there were 8 proteins found to be dysregulated differently between ET patients with mutated CALRdel52 and those with JAK2V617F mutation. These proteins might be related to the unique signaling pathways activated by CALRdel52 mutation which were different to JAK/STATs pathway by JAK2V617F mutation. Conclusion Our study demonstrated that numerous alterations of signaling proteins and pathways in ET patients with mutated CALRdel52. These findings could help to gain insights into the pathological mechanisms of ET.
Collapse
Affiliation(s)
- Wuhan Hui
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wei Zhang
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Congyan Liu
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Suigui Wan
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wanling Sun
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Li Su
- Department of Hematology, Xuan Wu Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
16
|
McKenna E, Mhaonaigh AU, Wubben R, Dwivedi A, Hurley T, Kelly LA, Stevenson NJ, Little MA, Molloy EJ. Neutrophils: Need for Standardized Nomenclature. Front Immunol 2021; 12:602963. [PMID: 33936029 PMCID: PMC8081893 DOI: 10.3389/fimmu.2021.602963] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Neutrophils are the most abundant innate immune cell with critical anti-microbial functions. Since the discovery of granulocytes at the end of the nineteenth century, the cells have been given many names including phagocytes, polymorphonuclear neutrophils (PMN), granulocytic myeloid derived suppressor cells (G-MDSC), low density neutrophils (LDN) and tumor associated neutrophils (TANS). This lack of standardized nomenclature for neutrophils suggest that biologically distinct populations of neutrophils exist, particularly in disease, when in fact these may simply be a manifestation of the plasticity of the neutrophil as opposed to unique populations. In this review, we profile the surface markers and granule expression of each stage of granulopoiesis to offer insight into how each stage of maturity may be identified. We also highlight the remarkable surface marker expression profiles between the supposed neutrophil populations.
Collapse
Affiliation(s)
- Ellen McKenna
- Discipline of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Paediatric Research Laboratory, Trinity Translational Medicine Institute (TTMI), St James' Hospital, Dublin, Ireland
| | | | - Richard Wubben
- Viral Immunology Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Amrita Dwivedi
- Trinity Health Kidney Centre, TTMI, Trinity College, Dublin, Ireland
| | - Tim Hurley
- Discipline of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Paediatric Research Laboratory, Trinity Translational Medicine Institute (TTMI), St James' Hospital, Dublin, Ireland.,Neonatology, Coombe Women and Infant's University Hospital, Dublin, Ireland
| | - Lynne A Kelly
- Discipline of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Paediatric Research Laboratory, Trinity Translational Medicine Institute (TTMI), St James' Hospital, Dublin, Ireland.,National Children's Research Centre, Dublin, Ireland
| | - Nigel J Stevenson
- Viral Immunology Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Dublin, Ireland.,Viral Immunology Group, Royal College of Surgeons in Ireland-Medical University of Bahrain, Zallaq, Bahrain
| | - Mark A Little
- Trinity Health Kidney Centre, TTMI, Trinity College, Dublin, Ireland.,Irish Centre for Vascular Biology, Trinity College Dublin, Dublin, Ireland
| | - Eleanor J Molloy
- Discipline of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Paediatric Research Laboratory, Trinity Translational Medicine Institute (TTMI), St James' Hospital, Dublin, Ireland.,Neonatology, Coombe Women and Infant's University Hospital, Dublin, Ireland.,National Children's Research Centre, Dublin, Ireland.,Neonatology, Children's Hospital Ireland (CHI) at Crumlin, Dublin, Ireland.,Paediatrics, CHI at Tallaght, Tallaght University Hospital, Dublin, Ireland
| |
Collapse
|
17
|
Immune complex-induced apoptosis and concurrent immune complex clearance are anti-inflammatory neutrophil functions. Cell Death Dis 2021; 12:296. [PMID: 33741905 PMCID: PMC7979711 DOI: 10.1038/s41419-021-03528-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 02/09/2021] [Accepted: 02/12/2021] [Indexed: 12/12/2022]
Abstract
Persistent neutrophilic inflammation drives host damage in autoimmune diseases that are characterized by abundant immune complexes. Insoluble immune complexes (iICs) potently activate pro-inflammatory neutrophil effector functions. We and others have shown that iICs also promote resolution of inflammation via stimulation of neutrophil apoptosis. We demonstrate here that iICs trigger FcγRIIa-dependent neutrophil macropinocytosis, leading to the rapid uptake, and subsequent degradation of iICs. We provide evidence that concurrent iIC-induced neutrophil apoptosis is distinct from phagocytosis-induced cell death. First, uptake of iICs occurs by FcγRII-stimulated macropinocytosis, rather than phagocytosis. Second, production of reactive oxygen species, but not iIC-internalization is a pre-requisite for iIC-induced neutrophil apoptosis. Our findings identify a previously unknown mechanism by which neutrophils can remove pro-inflammatory iICs from the circulation. Together iIC clearance and iIC-induced neutrophil apoptosis may act to prevent the potential escalation of neutrophilic inflammation in response to iICs.
Collapse
|
18
|
Traum A, Hofmann C, Haas S, Schmidt S, Bein G, Sachs UJ, Bayat B. Characterization of CD177-reactive iso- and auto-antibodies. Transfusion 2021; 61:1916-1922. [PMID: 33734454 DOI: 10.1111/trf.16359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND CD177 is a surface protein on neutrophils and a main mediator for the surface expression of proteinase 3 (PR3). Its functions are largely unknown. At least three types of antibodies have been described to target CD177: isoantibodies, which are formed in CD177-null individuals as a result of an immune reaction following transfusion or pregnancy; autoantibodies present in sera from patients with autoimmune neutropenia; and antineutrophil cytoplasmic antibodies in sera from patients with glomerulonephritis with polyangiitis. In this study, we aimed to compare the binding characteristics of auto- and iso-antibodies to optimize their detectability in the neutrophil serology laboratory. STUDY DESIGN AND METHODS The reactivity of iso- and auto-antibodies against CD177 was studied using granulocytes, "native" CD177/PR3 complex, and recombinant CD177 or PR3. RESULTS All iso- and auto-antibodies were reactive with CD177/PR3 when immobilized with monoclonal antibody (moab) 7D8. Seventy-five percent of autoantibodies, but none of the isoantibodies, did not react with CD177/PR3 immobilized with moab MEM166. The majority of autoantibodies did not react with recombinant CD177, whereas most isoantibodies tested positive. DISCUSSION Our results suggest that iso- and auto-antibodies against CD177 target different epitopes. Isoantibodies mainly target CD177 alone, while the majority of autoantibodies target a native epitope present on the neutrophil surface, but absent from recombinant CD177 which lacks PR3. Moab MEM166 binds to the native epitope and hinders the binding of CD177 autoantibodies. The results may help to design diagnostic strategies, especially for the identification of autoantibodies.
Collapse
Affiliation(s)
- Annalena Traum
- Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany
| | - Christine Hofmann
- Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany
| | - Sabine Haas
- Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany
| | - Silke Schmidt
- Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany
| | - Gregor Bein
- Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany
| | - Ulrich J Sachs
- Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany
| | - Behnaz Bayat
- Institute for Clinical Immunology and Transfusion Medicine, Justus Liebig University, Giessen, Germany
| |
Collapse
|
19
|
Filep JG, Ariel A. Neutrophil heterogeneity and fate in inflamed tissues: implications for the resolution of inflammation. Am J Physiol Cell Physiol 2020; 319:C510-C532. [PMID: 32667864 DOI: 10.1152/ajpcell.00181.2020] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Neutrophils are polymorphonuclear leukocytes that play a central role in host defense against infection and tissue injury. They are rapidly recruited to the inflamed site and execute a variety of functions to clear invading pathogens and damaged cells. However, many of their defense mechanisms are capable of inflicting collateral tissue damage. Neutrophil-driven inflammation is a unifying mechanism underlying many common diseases. Efficient removal of neutrophils from inflammatory loci is critical for timely resolution of inflammation and return to homeostasis. Accumulating evidence challenges the classical view that neutrophils represent a homogeneous population and that halting neutrophil influx is sufficient to explain their rapid decline within inflamed loci during the resolution of protective inflammation. Hence, understanding the mechanisms that govern neutrophil functions and their removal from the inflammatory locus is critical for minimizing damage to the surrounding tissue and for return to homeostasis. In this review, we briefly address recent advances in characterizing neutrophil phenotypic and functional heterogeneity and the molecular mechanisms that determine the fate of neutrophils within inflammatory loci and the outcome of the inflammatory response. We also discuss how these mechanisms may be harnessed as potential therapeutic targets to facilitate resolution of inflammation.
Collapse
Affiliation(s)
- János G Filep
- Department of Pathology and Cell Biology, University of Montreal and Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| | - Amiram Ariel
- Departmentof Biology and Human Biology, University of Haifa, Haifa, Israel
| |
Collapse
|
20
|
Yvan-Charvet L, Ng LG. Granulopoiesis and Neutrophil Homeostasis: A Metabolic, Daily Balancing Act. Trends Immunol 2020; 40:598-612. [PMID: 31256783 DOI: 10.1016/j.it.2019.05.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/06/2019] [Accepted: 05/06/2019] [Indexed: 02/07/2023]
Abstract
Granulopoiesis is part of the hematopoietic hierarchic architecture, where hematopoietic stem cells give rise to highly proliferative multipotent and lineage-committed granulocytic progenitor cells that differentiate into unipotent neutrophil progenitors. Given their short lifespan, neutrophils are rapidly cleared from circulation through specialized efferocytic macrophages. Together with an intrinsic clock, these processes contribute to circadian fluctuations, preserving self-tolerance and protection against invading pathogens. However, metabolic perturbation of granulopoiesis and neutrophil homeostasis can result in low-grade chronic inflammation, as observed with aging. During acute pathogenic infections, hematopoiesis can also be switched into emergency mode, which has been recently associated with significant neutrophil functional heterogeneity. This review focuses on a new reassessment of regulatory mechanisms governing neutrophil production, life-cycle, and diversity in health and disease.
Collapse
Affiliation(s)
- Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France.
| | - Lai Guan Ng
- Singapore Immunology Network (SIgN), A*STAR, Biopolis, Singapore 138648, Singapore; State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences, 288 Nanjing Road, Tianjin 300020, China; School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; Department of Microbiology & Immunology, Immunology Programme, Life Science Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
| |
Collapse
|
21
|
Dahlstrand Rudin A, Amirbeagi F, Davidsson L, Khamzeh A, Thorbert Mros S, Thulin P, Welin A, Björkman L, Christenson K, Bylund J. The neutrophil subset defined by CD177 expression is preferentially recruited to gingival crevicular fluid in periodontitis. J Leukoc Biol 2020; 109:349-362. [PMID: 32531826 DOI: 10.1002/jlb.3a0520-081rr] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/15/2020] [Accepted: 05/17/2020] [Indexed: 01/04/2023] Open
Abstract
In recent years, the concept of distinct subpopulations of human neutrophils has attracted much attention. One bona fide subset marker, exclusively expressed by a proportion of circulating neutrophils in a given individual, and therefore dividing neutrophils in two distinct subpopulations, is the glycoprotein CD177. CD177 is expressed on the plasma and granule membranes of 0-100% of circulating neutrophils depending on the donor. Several in vitro studies have linked CD177 to neutrophil transmigration, yet very few have looked at the role of CD177 for tissue recruitment in vivo. We investigate whether the CD177+ and CD177- neutrophil subsets differ in their propensity to migrate to both aseptic- and microbe-triggered inflamed human tissues. Microbe-triggered neutrophil migration was evaluated in samples of gingival crevicular fluid (GCF) from patients with periodontitis, whereas neutrophil migration to aseptic inflammation was evaluated in synovial fluid from patients with inflammatory arthritis, as well as in exudate from experimental skin chambers applied on healthy donors. We found that the proportion of CD177+ neutrophils was significantly higher in GCF from patients with periodontitis, as compared to blood from the same individuals. Such accumulation of CD177+ neutrophils was not seen in the two models of aseptic inflammation. Moreover, the proportion of CD177+ neutrophils in circulation was significantly higher in the periodontitis patient group, as compared to healthy donors. Our data indicate that the CD177+ neutrophil subset is preferentially recruited to the gingival crevice of periodontitis patients, and may imply that this subtype is of particular importance for situations of microbe-driven inflammation.
Collapse
Affiliation(s)
- Agnes Dahlstrand Rudin
- Department of Oral Microbiology and Immunology, Institute of Odontology. Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Firoozeh Amirbeagi
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Lisa Davidsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Arsham Khamzeh
- Department of Oral Microbiology and Immunology, Institute of Odontology. Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Sara Thorbert Mros
- Specialist Clinic of Periodontics, Gothenburg, Public Dental Service, Region Västra Götaland, Sweden
| | - Pontus Thulin
- Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Amanda Welin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Lena Björkman
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Unit of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Karin Christenson
- Department of Oral Microbiology and Immunology, Institute of Odontology. Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Johan Bylund
- Department of Oral Microbiology and Immunology, Institute of Odontology. Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
22
|
|
23
|
Calvente CJ, Tameda M, Johnson CD, del Pilar H, Lin YC, Adronikou N, De Mollerat Du Jeu X, Llorente C, Boyer J, Feldstein AE. Neutrophils contribute to spontaneous resolution of liver inflammation and fibrosis via microRNA-223. J Clin Invest 2019; 129:4091-4109. [PMID: 31295147 PMCID: PMC6763256 DOI: 10.1172/jci122258] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/03/2019] [Indexed: 02/06/2023] Open
Abstract
Persistent, unresolved inflammation in the liver represents a key trigger for hepatic injury and fibrosis in various liver diseases and is controlled by classically activated pro-inflammatory macrophages, while restorative macrophages of the liver are capable of reversing inflammation once the injury trigger ceases. Here we have identified a novel role for neutrophils as key contributors to resolving the inflammatory response in the liver. Using two models of liver inflammatory resolution, we found that mice undergoing neutrophil depletion during the resolution phase exhibited unresolved hepatic inflammation, activation of the fibrogenic machinery and early fibrosis. These findings were associated with an impairment of the phenotypic switch of pro-inflammatory macrophages into a restorative stage after removal of the cause of injury and an increased NLRP3 / miR-223 ratio. Mice with a deletion of the granulocyte specific miR-223 gene showed a similarly impaired resolution profile that could be reversed by restoring miR-223 levels using a miR-223 3p mimic or infusing neutrophils from wildtype animals. Collectively, our findings reveal a novel role for neutrophils in the liver as resolving effector cells that induce pro-inflammatory macrophages into a restorative phenotype, potentially via miR-223.
Collapse
Affiliation(s)
| | - Masahiko Tameda
- Department of Pediatrics, School of Medicine, UCSD, San Diego, California, USA
| | - Casey D. Johnson
- Department of Pediatrics, School of Medicine, UCSD, San Diego, California, USA
| | - Hana del Pilar
- Department of Pediatrics, School of Medicine, UCSD, San Diego, California, USA
| | - Yun Chin Lin
- Department of Pediatrics, School of Medicine, UCSD, San Diego, California, USA
| | | | | | - Cristina Llorente
- Department of Medicine, School of Medicine, UCSD, San Diego, California, USA
| | - Josh Boyer
- Department of Medicine, School of Medicine, UCSD, San Diego, California, USA
| | - Ariel E. Feldstein
- Department of Pediatrics, School of Medicine, UCSD, San Diego, California, USA
| |
Collapse
|
24
|
Bonaventura A, Montecucco F, Dallegri F, Carbone F, Lüscher TF, Camici GG, Liberale L. Novel findings in neutrophil biology and their impact on cardiovascular disease. Cardiovasc Res 2019; 115:1266-1285. [PMID: 30918936 DOI: 10.1093/cvr/cvz084] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa – Italian Cardiovascular Network, 10 Largo Benzi, Genoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa – Italian Cardiovascular Network, 10 Largo Benzi, Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
- Heart Division, Royal Brompton and Harefield Hospitals and Imperial College, London, UK
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
- University Heart Center, University Hospital Zürich, Rämistrasse 100, Zürich, Switzerland
- Department of Research and Education, University Hospital Zürich, Rämistrasse 100, Zürich, Switzerland
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
| |
Collapse
|
25
|
Grieshaber-Bouyer R, Nigrovic PA. Neutrophil Heterogeneity as Therapeutic Opportunity in Immune-Mediated Disease. Front Immunol 2019; 10:346. [PMID: 30886615 PMCID: PMC6409342 DOI: 10.3389/fimmu.2019.00346] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 02/11/2019] [Indexed: 12/25/2022] Open
Abstract
Neutrophils are versatile innate effector cells essential for immune defense but also responsible for pathologic inflammation. This dual role complicates therapeutic targeting. However, neither neutrophils themselves nor the mechanisms they employ in different forms of immune responses are homogeneous, offering possibilities for selective intervention. Here we review heterogeneity within the neutrophil population as well as in the pathways mediating neutrophil recruitment to inflamed tissues with a view to outlining opportunities for therapeutic manipulation in inflammatory disease.
Collapse
Affiliation(s)
- Ricardo Grieshaber-Bouyer
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA, United States
| | - Peter A Nigrovic
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA, United States.,Division of Immunology, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
26
|
Mortaz E, Alipoor SD, Adcock IM, Mumby S, Koenderman L. Update on Neutrophil Function in Severe Inflammation. Front Immunol 2018; 9:2171. [PMID: 30356867 PMCID: PMC6190891 DOI: 10.3389/fimmu.2018.02171] [Citation(s) in RCA: 245] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 09/03/2018] [Indexed: 12/17/2022] Open
Abstract
Neutrophils are main players in the effector phase of the host defense against micro-organisms and have a major role in the innate immune response. Neutrophils show phenotypic heterogeneity and functional flexibility, which highlight their importance in regulation of immune function. However, neutrophils can play a dual role and besides their antimicrobial function, deregulation of neutrophils and their hyperactivity can lead to tissue damage in severe inflammation or trauma. Neutrophils also have an important role in the modulation of the immune system in response to severe injury and trauma. In this review we will provide an overview of the current understanding of neutrophil subpopulations and their function during and post-infection and discuss the possible mechanisms of immune modulation by neutrophils in severe inflammation.
Collapse
Affiliation(s)
- Esmaeil Mortaz
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shamila D Alipoor
- Molecular Medicine Department, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Ian M Adcock
- Priority Research Centre for Asthma and Respiratory Disease, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia.,Airways Disease Section, Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Sharon Mumby
- Airways Disease Section, Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Leo Koenderman
- Laboratory of Translational Immunology, Department of Respiratory Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| |
Collapse
|
27
|
Korkmaz B, Caughey GH, Chapple I, Gauthier F, Hirschfeld J, Jenne DE, Kettritz R, Lalmanach G, Lamort AS, Lauritzen C, Łȩgowska M, Lesner A, Marchand-Adam S, McKaig SJ, Moss C, Pedersen J, Roberts H, Schreiber A, Seren S, Thakker NS. Therapeutic targeting of cathepsin C: from pathophysiology to treatment. Pharmacol Ther 2018; 190:202-236. [DOI: 10.1016/j.pharmthera.2018.05.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
28
|
Deng H, Hu N, Wang C, Chen M, Zhao MH. Interaction between CD177 and platelet endothelial cell adhesion molecule-1 downregulates membrane-bound proteinase-3 (PR3) expression on neutrophils and attenuates neutrophil activation induced by PR3-ANCA. Arthritis Res Ther 2018; 20:213. [PMID: 30236159 PMCID: PMC6148996 DOI: 10.1186/s13075-018-1710-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 08/28/2018] [Indexed: 01/07/2023] Open
Abstract
Background A recent study found that CD177 served as a receptor of membrane-bound proteinase-3 (mPR3) in a subset of neutrophils. Furthermore, CD177 has been identified as a high-affinity heterophilic binding partner for the endothelial cell platelet endothelial cell adhesion molecule-1 (PECAM-1). The current study aimed to investigate whether the interaction between PECAM-1 and CD177 could influence mPR3 expression as well as PR3-antineutrophil cytoplasmic antibody (ANCA)-induced neutrophil activation and glomerular endothelial cell (GEnC) injury. Methods The effect of interaction between CD177 and PECAM-1 on mPR3 expression was explored by enzyme-linked immunosorbent assay (ELISA) and flow cytometry. The effect of PECAM-1 on neutrophil activation and GEnC injury induced by PR3-ANCA-positive immunoglobulin (Ig)Gs was evaluated by dihydrorhodamine (DHR) assay and ELISA. CD177-negative neutrophils were selected by magnetic cell sorting (MACS), and the inhibitory effect of PECAM-1 on CD177-negative and mixed neutrophils was explored by measuring neutrophil degranulation. Results The level of specific interaction between CD177 and PECAM-1 was elevated with increasing CD177 concentration. The expression of mPR3 significantly decreased in neutrophils preincubated with PECAM-1 in a dose-dependent manner. Consistently, the levels of respiratory burst and degranulation induced by PR3-ANCA-positive IgGs in recombinant human tumor necrosis factor-alpha (TNF-α)-primed neutrophils was significantly reduced by preincubation with PECAM-1 (440.6 ± 123.0 vs. 511.4 ± 95.5, p < 0.05; and 3155.0 ± 1733.0 ng/ml vs. 5903.0 ± 717.5 ng/ml, p < 0.05, respectively). In CD177-negative neutrophils incubated with PR3-ANCA-positive IgGs, the level of degranulation was not significantly changed by preincubation with PECAM-1. However, in mixed neutrophils, PECAM-1 significantly decreased the level of degranulation induced by PR3-ANCA-positive IgGs (1015.9 ± 229.2% vs. 1725.2 ± 412.4%, p < 0.01). Furthermore, with preincubation of TNF-α-primed neutrophils with PECAM-1, the level of soluble intercellular cell adhesion molecule-1 (sICAM-1), a marker of endothelial cell activation and injury, in the supernatant of GEnCs treated with primed neutrophils plus PR3-ANCA-positive IgGs was significantly attenuated (112.7 ± 24.2 pg/ml vs. 167.5 ± 27.7 pg/ml, p < 0.05). Conclusions PECAM-1 can decrease the level of mPR3 expression on neutrophils, resulting in attenuation of neutrophil activation and subsequent GEnC injury induced by PR3-ANCA-positive IgGs.
Collapse
Affiliation(s)
- Hui Deng
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, 100034, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Peking University, Beijing, 100034, China.,Peking-Tsinghua Center for Life Sciences, Beijing, 100034, China
| | - Nan Hu
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, 100034, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Peking University, Beijing, 100034, China.,Peking-Tsinghua Center for Life Sciences, Beijing, 100034, China
| | - Chen Wang
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, 100034, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Peking University, Beijing, 100034, China.,Peking-Tsinghua Center for Life Sciences, Beijing, 100034, China
| | - Min Chen
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, 100034, China. .,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China. .,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Peking University, Beijing, 100034, China. .,Peking-Tsinghua Center for Life Sciences, Beijing, 100034, China.
| | - Ming-Hui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, 100034, China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, 100034, China.,Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education, Peking University, Beijing, 100034, China.,Peking-Tsinghua Center for Life Sciences, Beijing, 100034, China
| |
Collapse
|
29
|
Yang TH, St John LS, Garber HR, Kerros C, Ruisaard KE, Clise-Dwyer K, Alatrash G, Ma Q, Molldrem JJ. Membrane-Associated Proteinase 3 on Granulocytes and Acute Myeloid Leukemia Inhibits T Cell Proliferation. THE JOURNAL OF IMMUNOLOGY 2018; 201:1389-1399. [PMID: 30021768 DOI: 10.4049/jimmunol.1800324] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/24/2018] [Indexed: 11/19/2022]
Abstract
Proteinase 3 (P3), a serine protease expressed by myeloid cells, localized within azurophil granules, and also expressed on the cellular membrane of polymorphonuclear neutrophils (PMN), is the target of autoimmunity in granulomatosis with polyangiitis. PR1, an HLA-A2 restricted nonameric peptide derived from P3, has been targeted effectively in myeloid leukemia. We previously showed (Molldrem et al. 2003. JClinInvest 111: 639-647) that overexpression of P3 in chronic myeloid leukemia induces apoptosis of high-affinity PR1-specific T cells, leading to deletional tolerance and leukemia outgrowth. In this study, we investigated the effect of membrane P3 (mP3)-expressing PMN and acute myeloid leukemia (AML) blasts on the proliferation of CD4 and CD8 T cells in vitro. We demonstrate that mP3-expressing PMN significantly inhibits autologous healthy donor T cell proliferation but does not affect cytokine production in activated T cells and that this effect requires cell proximity and was abrogated by P3 blockade. This inhibition required P3 enzyme activity. However, suppression was not reversed by either the addition of catalase or the inhibition of arginase I. In addition to P3 blockade, anti-low density lipoprotein receptor-related protein 1 (LRP1) Ab also restored T cells' capacity to proliferate. Last, we show dose-dependent inhibition of T cell proliferation by mP3-expressing AML blasts. Together, our findings demonstrate a novel mechanism whereby PMN- and AML-associated mP3 inhibits T cell proliferation via direct LRP1 and mP3 interaction, and we identify P3 as a novel target to modulate immunity in myeloid leukemia and autoimmune disease.
Collapse
Affiliation(s)
- Tian-Hui Yang
- Section of Transplant Immunology, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Lisa S St John
- Section of Transplant Immunology, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Haven R Garber
- Section of Transplant Immunology, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Celine Kerros
- Section of Transplant Immunology, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Kathryn E Ruisaard
- Section of Transplant Immunology, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Karen Clise-Dwyer
- Section of Transplant Immunology, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Gheath Alatrash
- Section of Transplant Immunology, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Qing Ma
- Section of Transplant Immunology, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Jeffrey J Molldrem
- Section of Transplant Immunology, Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| |
Collapse
|
30
|
Miettinen HM, Gripentrog JM, Lord CI, Nagy JO. CD177-mediated nanoparticle targeting of human and mouse neutrophils. PLoS One 2018; 13:e0200444. [PMID: 29990379 PMCID: PMC6039027 DOI: 10.1371/journal.pone.0200444] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 06/26/2018] [Indexed: 12/23/2022] Open
Abstract
Neutrophils are the most abundant white blood cells, with a vital role in innate immune defense against bacterial and fungal pathogens. Although mostly associated with pathological processes directly related to immune defense, they can also play a detrimental role in inflammatory conditions and have been found to have a pro-metastatic role in the spread of cancer cells. Here, we explore ways to temporarily suppress these detrimental activities. We first examined the possibility of using siRNA and antisense oligonucleotides (ASOs) for transient knockdown of the human and mouse C5a receptor, an important chemoattractant receptor involved in neutrophil-mediated injury that is associated with myocardial infarction, sepsis, and neurodegenerative diseases. We found that siRNAs and ASOs transfected into cultured cell lines can eliminate 70–90% of C5a receptor mRNA and protein within 72 h of administration, a clinically relevant time frame after a cardiovascular event. Targeted drug delivery to specific cells or tissues of interest in a mammalian host, however, remains a major challenge. Here, using phage display technology, we have identified peptides that bind specifically to CD177, a neutrophil-specific surface molecule. We have attached these peptides to fluorescent, lipid-based nanoparticles and confirmed targeting and delivery to cultured cells ectopically presenting either human or mouse CD177. In addition, we have shown peptide-nanoparticle binding specifically to neutrophils in human and mouse blood. We anticipate that these or related tagged nanoparticles may be therapeutically useful for delivery of siRNAs or ASOs to neutrophils for transient knockdown of pro-inflammatory proteins such as the C5a receptor.
Collapse
Affiliation(s)
- Heini M. Miettinen
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States of America
- * E-mail:
| | - Jeannie M. Gripentrog
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States of America
| | - Connie I. Lord
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States of America
| | - Jon O. Nagy
- NanoValent Pharmaceuticals, Inc., Bozeman, MT, United States of America
| |
Collapse
|
31
|
Deniset JF, Kubes P. Neutrophil heterogeneity: Bona fide subsets or polarization states? J Leukoc Biol 2018; 103:829-838. [PMID: 29462505 DOI: 10.1002/jlb.3ri0917-361r] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 12/14/2022] Open
Abstract
Neutrophils are key components of the innate immune system that play important roles during infection, injury, and chronic disease. In recent years, neutrophil heterogeneity has become an emerging focus with accumulating evidence of neutrophil populations with distinct functions under both steady-state and pathologic conditions. Despite these advances, it remains unclear whether these different populations represent bona fide subsets or simply activation/polarization states in response to local cues. In this review, we summarize the varied neutrophils populations that have been described under both basal and during inflammation. We discuss the evidence that supports the existence of neutrophils subsets. Finally, we identify potential gaps in our knowledge that may further advance our current understanding of neutrophil heterogeneity.
Collapse
Affiliation(s)
- Justin F Deniset
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Paul Kubes
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.,Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
32
|
Abstract
As one of the first defenders of innate immune response, neutrophils make a rapid and robust response against infection or harmful agents. While traditionally regarded as suicidal killers that cause collateral tissue damage, recent findings on neutrophil extracellular trap formation, heterogeneity and plasticity and novel reparative functions have expanded our understanding of their diverse roles in health and disease. This review summarizes our current understanding of neutrophil-associated tissue injury, highlighting the emerging roles of neutrophil extracellular traps. This review will also focus on scrutinizing the roles of neutrophils in tissue repair and regeneration and will examine data on unexpected aspects of involvement of neutrophils in regulating normal tissue homeostasis.
Collapse
Affiliation(s)
- Jing Wang
- Division of Inflammation Biology, Institute of Enzyme Research, Tokushima University, Tokushima, Japan.
| |
Collapse
|
33
|
Liu H, Liu Y, Li Y, Liu Z, Li L, Ding S, Wang Y, Zhang T, Li L, Shao Z, Fu R. Proteinase 3 expression on the neutrophils of patients with paroxysmal nocturnal hemoglobinuria. Exp Ther Med 2017; 15:2525-2532. [PMID: 29467851 DOI: 10.3892/etm.2017.5662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 07/20/2017] [Indexed: 12/26/2022] Open
Abstract
Proteinase 3 (PR3) is released from neutrophils and regulates platelet activity, which is associated with cluster of differentiation (CD)177 antigen (NB1), a glycosylphosphatidylinositol-linked protein. In the present study, the effect of PR3 on thrombosis in paroxysmal nocturnal hemoglobinuria (PNH) and PNH-aplastic anemia (AA) syndrome was explored. The expression of PR3 and NB1 on CD59- neutrophils was detected by flow cytometry, immunofluorescence (IF), reverse transcription-quantitative polymerase chain reaction analysis and western blotting. Serum levels of PR3, proteinase-activated receptor 1 (PAR1) and D-Dimer were measured using ELISAs. The expression of PR3 and NB1 on the plasma membrane of CD59- neutrophils in patients with PNH/PNH-AA was significantly lower compared with their expression on CD59+ neutrophils in patients and controls (P=0.001). However, no correlation between PR3 and NB1 expression was identified. IF staining further demonstrated partially positive PR3 expression on CD59- neutrophils. The serum level of PR3 in patients was identified to be significantly decreased compared with healthy controls (P<0.0001), and significantly negatively correlated with PAR1 (r=-0.456; P=0.043) and D-Dimer (r=-0.503; P=0.028) levels. The mRNA and protein levels of PR3 on PNH clones did not change significantly compared with the control group. In conclusion, PR3 expression on the plasma membrane of neutrophils and in the serum of patients with PNH/PNH-AA decreased, which may result in increased PAR1 expression and increased clotting. The present study provides the basis for further study on platelets in PNH.
Collapse
Affiliation(s)
- Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yi Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yi Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Liyan Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Shaoxue Ding
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yihao Wang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Tian Zhang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Lijuan Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Zonghong Shao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
34
|
CD177 modulates human neutrophil migration through activation-mediated integrin and chemoreceptor regulation. Blood 2017; 130:2092-2100. [PMID: 28807980 DOI: 10.1182/blood-2017-03-768507] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 08/10/2017] [Indexed: 12/25/2022] Open
Abstract
CD177 is a glycosylphosphatidylinositol (GPI)-anchored protein expressed by a variable proportion of human neutrophils that mediates surface expression of the antineutrophil cytoplasmic antibody antigen proteinase 3. CD177 associates with β2 integrins and recognizes platelet endothelial cell adhesion molecule 1 (PECAM-1), suggesting a role in neutrophil migration. However, CD177pos neutrophils exhibit no clear migratory advantage in vivo, despite interruption of in vitro transendothelial migration by CD177 ligation. We sought to understand this paradox. Using a PECAM-1-independent transwell system, we found that CD177pos and CD177neg neutrophils migrated comparably. CD177 ligation selectively impaired migration of CD177pos neutrophils, an effect mediated through immobilization and cellular spreading on the transwell membrane. Correspondingly, CD177 ligation enhanced its interaction with β2 integrins, as revealed by fluorescence lifetime imaging microscopy, leading to integrin-mediated phosphorylation of Src and extracellular signal-regulated kinase (ERK). CD177-driven cell activation enhanced surface β2 integrin expression and affinity, impaired internalization of integrin attachments, and resulted in ERK-mediated attenuation of chemokine signaling. We conclude that CD177 signals in a β2 integrin-dependent manner to orchestrate a set of activation-mediated mechanisms that impair human neutrophil migration.
Collapse
|
35
|
Scapini P, Marini O, Tecchio C, Cassatella MA. Human neutrophils in the saga of cellular heterogeneity: insights and open questions. Immunol Rev 2017; 273:48-60. [PMID: 27558327 DOI: 10.1111/imr.12448] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent findings have uncovered novel fascinating aspects of the biology of neutrophils, which ultimately attribute to these cells a broader role in inflammation and immunity. One aspect that is currently under intensive investigation is the notion of neutrophil 'heterogeneity'. Studies examining neutrophils in a variety of acute and chronic inflammatory conditions report, in fact, the recovery of CD66b(+) cells displaying neutrophil-like morphology at different degrees of maturation/activation, able to exert either immunosuppressive or proinflammatory properties. These heterogeneous populations of mature and immature neutrophils are indicated with a variety of names, including 'low density neutrophils (LDNs)', 'low density granulocytes (LDGs)', 'granulocytic-myeloid derived suppressor cells (G-MDSCs)', and immunosuppressive neutrophils. However, due to the lack of discrete markers that can unequivocally allow their specific identification and isolation, the precise phenotype and function of all these presumably novel, neutrophil-like, populations have not been correctly defined yet. Aim of this article is to summarize current knowledge on the mature and immature neutrophil populations described to date, featuring immunosuppressive or proinflammatory properties, often defined as 'subsets', as well as to critically discuss unresolved issues in the field.
Collapse
Affiliation(s)
- Patrizia Scapini
- Division of General Pathology, University of Verona, Verona, Italy
| | - Olivia Marini
- Division of General Pathology, University of Verona, Verona, Italy
| | - Cristina Tecchio
- Division of Hematology and Bone Marrow Transplant Unit, University of Verona, Verona, Italy
| | | |
Collapse
|
36
|
Wada T, Miyamoto S, Okamoto H, Matsuda Y, Toma T, Imai K, Takagi M, Morio T, Yachie A. Prolonged neutropenia due to antihuman neutrophil antigen 2 (CD177) antibody after bone marrow transplantation. Pediatr Blood Cancer 2017; 64. [PMID: 27905683 DOI: 10.1002/pbc.26388] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/31/2016] [Accepted: 11/08/2016] [Indexed: 11/11/2022]
Abstract
We describe a patient who presented with prolonged neutropenia due to anti-human neutrophil antigen (HNA)-2 (CD177) antibody after allogeneic bone marrow transplantation. A granulocyte immunofluorescence test showed bimodal expression of antineutrophil antibody that resulted from specific binding of anti-HNA-2 to CD177+ neutrophils from healthy donors. The patient did not respond to granulocyte colony stimulating factor, which is able to upregulate CD177 expression on neutrophils. The low percentage of CD177+ cells in the few remaining neutrophils supports the possibility of elimination of CD177-upregulated neutrophils. These findings might explain an inferior response to neutrophil growth factors in neutropenia secondary to anti-HNA-2 antibody.
Collapse
Affiliation(s)
- Taizo Wada
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Satoshi Miyamoto
- Department of Pediatrics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Okamoto
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yusuke Matsuda
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Tomoko Toma
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Kohsuke Imai
- Department of Pediatrics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masatoshi Takagi
- Department of Pediatrics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomohiro Morio
- Department of Pediatrics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akihiro Yachie
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
37
|
Neutrophil migration in infection and wound repair: going forward in reverse. Nat Rev Immunol 2017; 16:378-91. [PMID: 27231052 DOI: 10.1038/nri.2016.49] [Citation(s) in RCA: 674] [Impact Index Per Article: 96.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neutrophil migration and its role during inflammation has been the focus of increased interest in the past decade. Advances in live imaging and the use of new model systems have helped to uncover the behaviour of neutrophils in injured and infected tissues. Although neutrophils were considered to be short-lived effector cells that undergo apoptosis in damaged tissues, recent evidence suggests that neutrophil behaviour is more complex and, in some settings, neutrophils might leave sites of tissue injury and migrate back into the vasculature. The role of reverse migration and its contribution to resolution of inflammation remains unclear. In this Review, we discuss the different cues within tissues that mediate neutrophil forward and reverse migration in response to injury or infection and the implications of these mechanisms to human disease.
Collapse
|
38
|
Garley M, Jabłońska E. Heterogeneity Among Neutrophils. Arch Immunol Ther Exp (Warsz) 2017; 66:21-30. [PMID: 28560557 PMCID: PMC5767199 DOI: 10.1007/s00005-017-0476-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 04/03/2017] [Indexed: 12/18/2022]
Abstract
Neutrophils (PMNs) play a key role in innate defence mechanisms. Generally, PMNs were considered to have a homogeneous population of mature and diversified cells. It seems, however, that their pleiotropic action results from the existence of different subpopulations in this group of cells. There are data that confirm the involvement of PMNs in the direct activation of other cells in non-specific response, as well as specialised cells in specific response. For example, there have been observations of PMNs with different levels of activity in relation to lymphocytes, and a population was identified which had characteristics similar to those of cells which are capable of presenting antigens. There are also reports of PMNs which demonstrate different survival time or capacity for chemotaxis. Other studies suggest that the neutrophil response to Staphylococcus aureus is diverse (not identical among all neutrophil). There are also reports of PMNs with varying activity during inflammation, which might explain many as yet unknown pathophysiological aspects of their hyperreactivity. The functional dualism of PMNs in the course of neoplastic disorders raises a lot of controversy. This paper presents the current state of knowledge of the heterogeneity of PMNs and their potential roles in different stages of disease.
Collapse
Affiliation(s)
- Marzena Garley
- Department of Immunology, Medical University of Bialystok, J. Waszyngtona 15A, 15-269, Białystok, Poland.
| | - Ewa Jabłońska
- Department of Immunology, Medical University of Bialystok, J. Waszyngtona 15A, 15-269, Białystok, Poland
| |
Collapse
|
39
|
Eulenberg-Gustavus C, Bähring S, Maass PG, Luft FC, Kettritz R. Gene silencing and a novel monoallelic expression pattern in distinct CD177 neutrophil subsets. J Exp Med 2017; 214:2089-2101. [PMID: 28559244 PMCID: PMC5502425 DOI: 10.1084/jem.20161093] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 02/01/2017] [Accepted: 04/12/2017] [Indexed: 12/24/2022] Open
Abstract
CD177 presents antigens in allo- and autoimmune diseases on the neutrophil surface. Eulenberg-Gustavus et al. show that epigenetic silencing causes CD177negative neutrophils, whereas a novel pattern of monoallelic CD177 expression results in a variable percentage of CD177positive neutrophils in bimodal individuals. CD177 presents antigens in allo- and autoimmune diseases on the neutrophil surface. Individuals can be either CD177-deficient or harbor distinct CD177neg and CD177pos neutrophil subsets. We studied mechanisms controlling subset-restricted CD177 expression in bimodal individuals. CD177pos, but not CD177neg neutrophils, produced CD177 protein and mRNA. Haplotype analysis indicated a unique monoallelic CD177 expression pattern, where the offspring stably transcribed either the maternal or paternal allele. Hematopoietic stem cells expressed both CD177 alleles and silenced one copy during neutrophil differentiation. ChIP and reporter assays in HeLa cells with monoallelic CD177 expression showed that methylation reduced reporter activity, whereas demethylation caused biallelic CD177 expression. HeLa cell transfection with c-Jun and c-Fos increased CD177 mRNA. Importantly, CD177pos human neutrophils, but not CD177neg neutrophils, showed a euchromatic CD177 promoter, unmethylated CpGs, and c-Jun and c-Fos binding. We describe epigenetic mechanisms explaining the two distinct CD177 neutrophil subsets and a novel monoallelic CD177 expression pattern that does not follow classical random monoallelic expression or imprinting.
Collapse
Affiliation(s)
- Claudia Eulenberg-Gustavus
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine at the Charité, Berlin, Germany
| | - Sylvia Bähring
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine at the Charité, Berlin, Germany
| | - Philipp G Maass
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
| | - Friedrich C Luft
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine at the Charité, Berlin, Germany
| | - Ralph Kettritz
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrück Center for Molecular Medicine at the Charité, Berlin, Germany .,Nephrology and Intensive Care Medicine, Campus Virchow, Medical Faculty of the Charité, Berlin, Germany
| |
Collapse
|
40
|
Martin KR, Witko-Sarsat V. Proteinase 3: the odd one out that became an autoantigen. J Leukoc Biol 2017; 102:689-698. [PMID: 28546501 DOI: 10.1189/jlb.3mr0217-069r] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/07/2017] [Accepted: 04/16/2017] [Indexed: 01/09/2023] Open
Abstract
Neutrophils are critical in the defense against bacterial and fungal pathogens, and they also modulate the inflammatory process. The areas where neutrophils are studied have expanded from the restricted field of antibacterial defense to the modulation of inflammation and finally, to fine-tuning immune responses. As a result, recent studies have shown that neutrophils are implicated in several systemic autoimmune diseases, although exactly how neutrophils contribute to these diseases and the molecular mechanisms responsible are still under investigation. In a group of autoimmune vasculitides associated with anti-neutrophil cytoplasmic antibodies (AAVs), granulomatosis with polyangiitis (GPA) illustrates the concept that autoimmunity can develop against one specific neutrophil protein, namely, proteinase 3 (PR3), one of the four serine protease homologs contained within azurophilic granules. In this review, we will focus on recent molecular analyses combined with functional studies that provide clear evidence that the pathogenic properties of PR3 are not only a result of its enzymatic activity but also mediated by a particular structural element-the hydrophobic patch-which facilitates associations with various proteins and lipids and permits anchorage into the plasma membrane. Furthermore, these unique structural and functional characteristics of PR3 might be key contributors to the systemic inflammation and to the immune dysregulation observed in GPA.
Collapse
Affiliation(s)
- Katherine R Martin
- Institut National de la Santé et de la Recherche Médicale, U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique-Unité Mixte de Recherche 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France; and.,Center of Excellence, LabEx Inflamex, Paris, France
| | - Véronique Witko-Sarsat
- Institut National de la Santé et de la Recherche Médicale, U1016, Institut Cochin, Paris, France; .,Centre National de la Recherche Scientifique-Unité Mixte de Recherche 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France; and.,Center of Excellence, LabEx Inflamex, Paris, France
| |
Collapse
|
41
|
Jimenez-Valdes RJ, Rodriguez-Moncayo R, Cedillo-Alcantar DF, Garcia-Cordero JL. Massive Parallel Analysis of Single Cells in an Integrated Microfluidic Platform. Anal Chem 2017; 89:5210-5220. [PMID: 28406613 DOI: 10.1021/acs.analchem.6b04485] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
New tools that facilitate the study of cell-to-cell variability could help uncover novel cellular regulation mechanisms. We present an integrated microfluidic platform to analyze a large number of single cells in parallel. To isolate and analyze thousands of individual cells in multiplexed conditions, our platform incorporates arrays of microwells (7 pL each) in a multilayered microfluidic device. The device allows the simultaneous loading of cells into 16 separate chambers, each containing 4640 microwells, for a total of 74 240 wells per device. We characterized different parameters important for the operation of the microfluidic device including flow rate, solution exchange rate in a microchamber, shear stress, and time to fill up a single microwell with molecules of different molecular weight. In general, after ∼7.5 min of cell loading our device has an 80% microwell occupancy with 1-4 cells, of which 36% of wells contained a single cell. To test the functionality of our device, we carried out a cell viability assay with adherent and nonadherent cells. We also studied the production of neutrophil extracellular traps (NETs) from single neutrophils isolated from peripheral blood, observing the existence of temporal heterogeneity in NETs production, perhaps having implications in the type of the neutrophil response to an infection or inflammation. We foresee our platform will have a variety of applications in drug discovery and cellular biology by facilitating the characterization of phenotypic differences in a monoclonal cell population.
Collapse
Affiliation(s)
- Rocio J Jimenez-Valdes
- Unidad Monterrey, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Via del Conocimiento 201, Parque PIIT, Apodaca, Nuevo León CP 66628, Mexico
| | - Roberto Rodriguez-Moncayo
- Unidad Monterrey, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Via del Conocimiento 201, Parque PIIT, Apodaca, Nuevo León CP 66628, Mexico
| | - Diana F Cedillo-Alcantar
- Unidad Monterrey, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Via del Conocimiento 201, Parque PIIT, Apodaca, Nuevo León CP 66628, Mexico
| | - Jose L Garcia-Cordero
- Unidad Monterrey, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional , Via del Conocimiento 201, Parque PIIT, Apodaca, Nuevo León CP 66628, Mexico
| |
Collapse
|
42
|
Characterization of the CD177 interaction with the ANCA antigen proteinase 3. Sci Rep 2017; 7:43328. [PMID: 28240246 PMCID: PMC5327412 DOI: 10.1038/srep43328] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 01/24/2017] [Indexed: 01/13/2023] Open
Abstract
Proteinase 3 is a serine protease found in neutrophil granules and on the extracellular neutrophil membrane (mPR3). mPR3 is a major antigen for anti-neutrophil cytoplasmic antibodies (PR3-ANCAs), autoantibodies causing fatal autoimmune diseases. In most individuals, a subpopulation of neutrophils also produce CD177, proposed to present additional PR3 on the surface, resulting in CD177neg/mPR3low and CD177pos/mPR3high neutrophil subsets. A positive correlation has been shown between mPR3 abundance, disease incidence, and clinical outcome. We present here a detailed investigation of the PR3:CD177 complex, verifying the interaction, demonstrating the effect of binding on PR3 proteolytic activity and explaining the accessibility of major PR3-ANCA epitopes. We observed high affinity PR3:CD177 complex formation by surface plasmon resonance. Using flow cytometry and a PR3-specific FRET assay, we found that CD177 binding reduced the proteolytic activity of PR3 in vitro using purified proteins, in neutrophil degranulation supernatants containing wtPR3 and directly on mPR3high neutrophils and PR3-loaded HEK cells. Finally, CD177pos/mPR3high neutrophils showed no migration advantage in vitro or in vivo when migrating from the blood into the oral cavity. We illuminate details of the PR3:CD177 interaction explaining mPR3 membrane orientation and proteolytic activity with relevance to ANCA activation of the distinct mPR3 neutrophil populations.
Collapse
|
43
|
Abstract
Neutrophils have long been regarded as key effectors of the innate immune response during acute inflammation. Recent evidence has revealed a greater functional diversity for these cells than previously appreciated, expanding roles for neutrophils in adaptive immunity and chronic pathologies. In this review, we summarize some of the evolving paradigms in the neutrophil field and highlight key advances that have contributed to our understanding of neutrophil behavior and function
in vivo. We examine the concept of neutrophil subsets and polarization, we discuss novel immunomodulatory roles for neutrophils in shaping the immune response, and, finally, we identify technical advances that will further enhance our ability to track the function and fate of neutrophils.
Collapse
Affiliation(s)
- Justin F Deniset
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada; Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada; Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
44
|
Affiliation(s)
- Ralph Kettritz
- Experimental and Clinical Research Center; A joint cooperation between the Charité and the Max-Delbrück Center for Molecular Medicine (MDC) and Department of Nephrology and Intensive Care Medicine; Charité University Health Services; Berlin Germany
| |
Collapse
|
45
|
Schreiber A, Eulenberg-Gustavus C, Bergmann A, Jerke U, Kettritz R. Lessons from a double-transgenic neutrophil approach to induce antiproteinase 3 antibody-mediated vasculitis in mice. J Leukoc Biol 2016; 100:1443-1452. [PMID: 27365530 DOI: 10.1189/jlb.5a0116-037r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/25/2016] [Accepted: 06/15/2016] [Indexed: 12/17/2022] Open
Abstract
ANCA to either PR3 or MPO are found in patients with necrotizing vasculitis and glomerulonephritis. ANCA binding to their target antigens on neutrophils and subsequent neutrophil activation are pivotal disease mechanisms that lead to vascular inflammation and necrosis. ANCA interaction with PR3 is more complex than with MPO as the neutrophil-specific CD177 receptor is involved in PR3 surface expression and PR3-ANCA-induced neutrophil activation. Modeling human disease is important to clinical research. Highly successful mouse models of MPO-ANCA vasculitis exist; however, recapitulating PR3-ANCA vasculitis has not been successful. We generated double-transgenic (DT) mice that expressed human PR3 and CD177 under a myeloid-specific huMRP8 promoter in an attempt to model PR3-ANCA vasculitis. DT mice strongly expressed the human transgenes in and on murine neutrophils and bound murine and human anti-PR3 antibodies. Nevertheless, passive transfer of these antibodies into LPS-primed DT mice or immunization of C57BL/6 mice with human PR3 followed by irradiation and transplantation of DT bone marrow failed to induce glomerulonephritis. Further analyses revealed that anti-PR3 antibodies did not activate DT neutrophils as shown by superoxide generation. Moreover, we found that mice did not properly process human pro-PR3 into mature PR3 and, consequently, the signaling complex between PR3, CD177, and CD11b, which promotes neutrophil activation by anti-PR3 antibodies, failed to form. We conclude that important species differences in PR3 and CD177 exist between men and mice that prevented successful generation of a murine anti-PR3 antibody model.
Collapse
Affiliation(s)
- Adrian Schreiber
- Experimental and Clinical Research Center, Charité, Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Nephrology and Intensive Care Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Eulenberg-Gustavus
- Experimental and Clinical Research Center, Charité, Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Astrid Bergmann
- Experimental and Clinical Research Center, Charité, Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Uwe Jerke
- Experimental and Clinical Research Center, Charité, Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ralph Kettritz
- Experimental and Clinical Research Center, Charité, Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; .,Nephrology and Intensive Care Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
46
|
Wu Z, Liang R, Ohnesorg T, Cho V, Lam W, Abhayaratna WP, Gatenby PA, Perera C, Zhang Y, Whittle B, Sinclair A, Goodnow CC, Field M, Andrews TD, Cook MC. Heterogeneity of Human Neutrophil CD177 Expression Results from CD177P1 Pseudogene Conversion. PLoS Genet 2016; 12:e1006067. [PMID: 27227454 PMCID: PMC4882059 DOI: 10.1371/journal.pgen.1006067] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 04/28/2016] [Indexed: 12/16/2022] Open
Abstract
Most humans harbor both CD177neg and CD177pos neutrophils but 1-10% of people are CD177null, placing them at risk for formation of anti-neutrophil antibodies that can cause transfusion-related acute lung injury and neonatal alloimmune neutropenia. By deep sequencing the CD177 locus, we catalogued CD177 single nucleotide variants and identified a novel stop codon in CD177null individuals arising from a single base substitution in exon 7. This is not a mutation in CD177 itself, rather the CD177null phenotype arises when exon 7 of CD177 is supplied entirely by the CD177 pseudogene (CD177P1), which appears to have resulted from allelic gene conversion. In CD177 expressing individuals the CD177 locus contains both CD177P1 and CD177 sequences. The proportion of CD177hi neutrophils in the blood is a heritable trait. Abundance of CD177hi neutrophils correlates with homozygosity for CD177 reference allele, while heterozygosity for ectopic CD177P1 gene conversion correlates with increased CD177neg neutrophils, in which both CD177P1 partially incorporated allele and paired intact CD177 allele are transcribed. Human neutrophil heterogeneity for CD177 expression arises by ectopic allelic conversion. Resolution of the genetic basis of CD177null phenotype identifies a method for screening for individuals at risk of CD177 isoimmunisation.
Collapse
Affiliation(s)
- Zuopeng Wu
- Translational Research Unit, Canberra Hospital, Woden, Australian Capital Territory, Australia
- Clinical Trials Unit, Canberra Hospital, Woden, Australian Capital Territory, Australia
| | - Rong Liang
- Australian Phenomics Facility, Australian National University, Australian Capital Territory, Australia
| | - Thomas Ohnesorg
- Murdoch Children’s Research Institute, Department of Paediatrics, The University of Melbourne, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Vicky Cho
- Department of Immunology, The John Curtin School of Medical Research, Australian National University, Acton, Australian Capital Territory, Australia
| | - Wesley Lam
- Translational Research Unit, Canberra Hospital, Woden, Australian Capital Territory, Australia
| | - Walter P. Abhayaratna
- Clinical Trials Unit, Canberra Hospital, Woden, Australian Capital Territory, Australia
| | - Paul A. Gatenby
- Department of Immunology, Canberra Hospital, Woden, Australian Capital Territory, Australia
| | - Chandima Perera
- Department of Rheumatology, Canberra Hospital, Woden, Australian Capital Territory, Australia
| | - Yafei Zhang
- Australian Phenomics Facility, Australian National University, Australian Capital Territory, Australia
| | - Belinda Whittle
- Australian Phenomics Facility, Australian National University, Australian Capital Territory, Australia
| | - Andrew Sinclair
- Murdoch Children’s Research Institute, Department of Paediatrics, The University of Melbourne, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Christopher C. Goodnow
- Department of Immunology, The John Curtin School of Medical Research, Australian National University, Acton, Australian Capital Territory, Australia
| | - Matthew Field
- Department of Immunology, The John Curtin School of Medical Research, Australian National University, Acton, Australian Capital Territory, Australia
| | - T. Daniel Andrews
- Department of Immunology, The John Curtin School of Medical Research, Australian National University, Acton, Australian Capital Territory, Australia
| | - Matthew C. Cook
- Translational Research Unit, Canberra Hospital, Woden, Australian Capital Territory, Australia
- Department of Immunology, The John Curtin School of Medical Research, Australian National University, Acton, Australian Capital Territory, Australia
- Department of Immunology, Canberra Hospital, Woden, Australian Capital Territory, Australia
- * E-mail:
| |
Collapse
|
47
|
Neutrophil heterogeneity: implications for homeostasis and pathogenesis. Blood 2016; 127:2173-81. [DOI: 10.1182/blood-2016-01-688887] [Citation(s) in RCA: 247] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 03/16/2016] [Indexed: 02/06/2023] Open
Abstract
Abstract
Neutrophils are polymorphonuclear leukocytes of the phagocytic system that act as first line of host defense against invading pathogens but are also important mediators of inflammation-induced injury. In contrast to other members of the innate immune system, neutrophils are classically considered a homogenous population of terminally differentiated cells with a well-defined and highly conserved function. Indeed, their short lifespan, the absent proliferative capacity, their limited ability to produce large amounts of cytokines, and the failure to recirculate from the tissue to the bloodstream have sustained this idea. However, increasing evidence over the last decade has demonstrated an unexpected phenotypic heterogeneity and functional versatility of the neutrophil population. Far beyond their antimicrobial functions, neutrophils are emerging as decision-shapers during innate and adaptive immune responses. These emerging discoveries open a new door to understand the role of neutrophils during homeostatic but also pathogenic immune processes. Thus, this review details novel insights of neutrophil phenotypic and functional heterogeneity during homeostasis and disease.
Collapse
|
48
|
Johansson ÅC, Ohlsson S, Pettersson Å, Bengtsson AA, Selga D, Hansson M, Hellmark T. Impaired phagocytosis and reactive oxygen species production in phagocytes is associated with systemic vasculitis. Arthritis Res Ther 2016; 18:92. [PMID: 27102815 PMCID: PMC4840900 DOI: 10.1186/s13075-016-0994-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/11/2016] [Indexed: 11/10/2022] Open
Abstract
Background Anti-neutrophil cytoplasmic antibodies associated vasculitides (AAV) is a group of autoimmune diseases, characterized by small vessel inflammation. Phagocytes such as neutrophils and monocytes are the main effector cells found around the inflamed vessel wall. Therefore, we wanted to investigate aspects of function and activation of these cells in patients with AAV. Methods Flow cytometry was used to evaluate: the expression of activation markers (CD11c, CD62L, CD177 and C5aR); the number of recently released neutrophils from bone marrow, defined as CD10-D16low cells in peripheral blood; and the capacity of peripheral blood monocytes and polymorphonuclear leukocytes (PMN) to produce reactive oxygen species and to phagocytose opsonized bacteria. Results AAV patients (n = 104) showed an increase of CD10-CD16low neutrophils and total PMN in peripheral blood, suggesting a combination of increased bone marrow release and prolonged survival. An increased percentage of AAV PMN expressed CD177 but no other signs of activation were seen. A decreased production of reactive oxygen species was observed in AAV phagocytes, which was associated with disease activity. Moreover, granulocytes from patients with microscopic polyangiitis showed lower oxidative burst capacity compared to patients with granulomatosis with polyangiitis or eosinophilic granulomatosis with polyangiitis. In addition, decreased phagocytosis capacity was seen in PMN and monocytes. Conclusion Our results indicate that phagocytes from AAV patients have impaired function, are easily mobilized from bone marrow but are not particularly activated. The association between low reactive oxygen species formation in PMN and disease severity is consistent with findings in other autoimmune diseases and might be considered as a risk factor. Electronic supplementary material The online version of this article (doi:10.1186/s13075-016-0994-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Åsa Cm Johansson
- Department of Haematology, Lund University and Skåne University Hospital, BMC B13, 221 84, Lund, Sweden. .,University and Regional Laboratories Region Skåne, Clinical Immunology and Transfusion Medicine, Skåne, 221 85, Lund, Sweden.
| | - Sophie Ohlsson
- Department of Clinical Sciences Lund, Nephrology, Lund University, Skane University Hospital, Lund, Sweden
| | - Åsa Pettersson
- Department of Clinical Sciences Lund, Nephrology, Lund University, Skane University Hospital, Lund, Sweden
| | - Anders A Bengtsson
- Department of Clinical Sciences, Lund, Rheumatology, Lund University, Skåne University Hospital, Lund, Sweden
| | - Daina Selga
- Department of Clinical Sciences Lund, Nephrology, Lund University, Skane University Hospital, Lund, Sweden
| | - Markus Hansson
- Department of Haematology, Lund University and Skåne University Hospital, BMC B13, 221 84, Lund, Sweden
| | - Thomas Hellmark
- Department of Clinical Sciences Lund, Nephrology, Lund University, Skane University Hospital, Lund, Sweden
| |
Collapse
|
49
|
Organization, evolution and functions of the human and mouse Ly6/uPAR family genes. Hum Genomics 2016; 10:10. [PMID: 27098205 PMCID: PMC4839075 DOI: 10.1186/s40246-016-0074-2] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/14/2016] [Indexed: 01/08/2023] Open
Abstract
Members of the lymphocyte antigen-6 (Ly6)/urokinase-type plasminogen activator receptor (uPAR) superfamily of proteins are cysteine-rich proteins characterized by a distinct disulfide bridge pattern that creates the three-finger Ly6/uPAR (LU) domain. Although the Ly6/uPAR family proteins share a common structure, their expression patterns and functions vary. To date, 35 human and 61 mouse Ly6/uPAR family members have been identified. Based on their subcellular localization, these proteins are further classified as GPI-anchored on the cell membrane, or secreted. The genes encoding Ly6/uPAR family proteins are conserved across different species and are clustered in syntenic regions on human chromosomes 8, 19, 6 and 11, and mouse Chromosomes 15, 7, 17, and 9, respectively. Here, we review the human and mouse Ly6/uPAR family gene and protein structure and genomic organization, expression, functions, and evolution, and introduce new names for novel family members.
Collapse
|
50
|
Thieblemont N, Wright HL, Edwards SW, Witko-Sarsat V. Human neutrophils in auto-immunity. Semin Immunol 2016; 28:159-73. [DOI: 10.1016/j.smim.2016.03.004] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 03/08/2016] [Accepted: 03/12/2016] [Indexed: 01/06/2023]
|