1
|
Frampton S, Smith R, Ferson L, Gibson J, Hollox EJ, Cragg MS, Strefford JC. Fc gamma receptors: Their evolution, genomic architecture, genetic variation, and impact on human disease. Immunol Rev 2024. [PMID: 39345014 DOI: 10.1111/imr.13401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Fc gamma receptors (FcγRs) are a family of receptors that bind IgG antibodies and interface at the junction of humoral and innate immunity. Precise regulation of receptor expression provides the necessary balance to achieve healthy immune homeostasis by establishing an appropriate immune threshold to limit autoimmunity but respond effectively to infection. The underlying genetics of the FCGR gene family are central to achieving this immune threshold by regulating affinity for IgG, signaling efficacy, and receptor expression. The FCGR gene locus was duplicated during evolution, retaining very high homology and resulting in a genomic region that is technically difficult to study. Here, we review the recent evolution of the gene family in mammals, its complexity and variation through copy number variation and single-nucleotide polymorphism, and impact of these on disease incidence, resolution, and therapeutic antibody efficacy. We also discuss the progress and limitations of current approaches to study the region and emphasize how new genomics technologies will likely resolve much of the current confusion in the field. This will lead to definitive conclusions on the impact of genetic variation within the FCGR gene locus on immune function and disease.
Collapse
Affiliation(s)
- Sarah Frampton
- Cancer Genomics Group, Faculty of Medicine, School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Rosanna Smith
- Antibody and Vaccine Group, Faculty of Medicine, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton, Southampton, UK
| | - Lili Ferson
- Cancer Genomics Group, Faculty of Medicine, School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Jane Gibson
- Cancer Genomics Group, Faculty of Medicine, School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Edward J Hollox
- Department of Genetics, Genomics and Cancer Sciences, College of Life Sciences, University of Leicester, Leicester, UK
| | - Mark S Cragg
- Antibody and Vaccine Group, Faculty of Medicine, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton, Southampton, UK
| | - Jonathan C Strefford
- Cancer Genomics Group, Faculty of Medicine, School of Cancer Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
2
|
Blanton LV, San Roman AK, Wood G, Buscetta A, Banks N, Skaletsky H, Godfrey AK, Pham TT, Hughes JF, Brown LG, Kruszka P, Lin AE, Kastner DL, Muenke M, Page DC. Stable and robust Xi and Y transcriptomes drive cell-type-specific autosomal and Xa responses in vivo and in vitro in four human cell types. CELL GENOMICS 2024; 4:100628. [PMID: 39111319 PMCID: PMC11480847 DOI: 10.1016/j.xgen.2024.100628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/11/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024]
Abstract
Recent in vitro studies of human sex chromosome aneuploidy showed that the Xi ("inactive" X) and Y chromosomes broadly modulate autosomal and Xa ("active" X) gene expression. We tested these findings in vivo. Linear modeling of CD4+ T cells and monocytes from individuals with one to three X chromosomes and zero to two Y chromosomes revealed 82 sex-chromosomal and 344 autosomal genes whose expression changed significantly with Xi and/or Y dosage in vivo. Changes in sex-chromosomal expression were remarkably constant in vivo and in vitro; autosomal responses to Xi and/or Y dosage were largely cell-type specific (∼2.6-fold more variation than sex-chromosomal responses). Targets of the sex-chromosomal transcription factors ZFX and ZFY accounted for a significant fraction of these autosomal responses both in vivo and in vitro. We conclude that the human Xi and Y transcriptomes are surprisingly robust and stable, yet they modulate autosomal and Xa genes in a cell-type-specific fashion.
Collapse
Affiliation(s)
| | | | - Geryl Wood
- Inflammatory Disease Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ashley Buscetta
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole Banks
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA; Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Helen Skaletsky
- Whitehead Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142, USA
| | | | - Thao T Pham
- Whitehead Institute, Cambridge, MA 02142, USA
| | | | - Laura G Brown
- Whitehead Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142, USA
| | - Paul Kruszka
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Angela E Lin
- Genetics Unit, MassGeneral for Children, Boston, MA 02114, USA
| | - Daniel L Kastner
- Inflammatory Disease Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Maximilian Muenke
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David C Page
- Whitehead Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
3
|
Wu Y, Zhou Y, Zhu Q, Liu Y, Deng D, Zhang J. B cell function in patients with systemic lupus erythematosus is regulated by the upregulation of JunD. Heliyon 2024; 10:e35949. [PMID: 39220919 PMCID: PMC11365431 DOI: 10.1016/j.heliyon.2024.e35949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Purpose Systemic lupus erythematosus (SLE) is largely caused by B cell dysfunction. JunD is an activator protein 1 family protein that has been linked to the regulation of apoptotic and proliferative activities. However, the precise mechanism(s) by which JunD functions remains to be fully elucidated. Accordingly, this study aimed to clarify the functional importance of JUND gene expression in SLE, with further analyses of the functional role that JunD plays as a regulator of B cell proliferation and immune function. Methods Reverse transcriptase quantitative polymerase chain reaction techniques were used to analyze JunD expression in B cells of patients with SLE and healthy subjects. Cell Counting Kit-8 (CCK-8) assays and flow cytometry methods were used to characterise proliferative activity, cell cycle progression, and apoptosis of B cells in which JunD was either knocked down or overexpressed. The immune status and autophagic activity of these cells were assessed using Western immunoblotting and enzyme-linked immunosorbent assay (ELISA). Additionally, a JunD knockdown mouse model was established, and the functional role of B cell JunD expression in the pathogenesis of SLE was assessed using Western immunoblotting, ELISA, and haematoxylin and eosin staining. Results B cells from patients with SLE exhibited upregulation of JunD, with overexpression facilitating in vitro cellular proliferation and modulation of the immune and autophagic status of these B cells. JunD knockdown was also sufficient to modulate in vivo immune function and the autophagic status of B cells. Conclusion JunD was upregulated in the B cells of patients with SLE, where it regulates proliferation, autophagy, and immunity.
Collapse
Affiliation(s)
- Yongzhuo Wu
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, PR China
| | - Yali Zhou
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, PR China
| | - Qinghuan Zhu
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, PR China
| | - Yingying Liu
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, PR China
| | - Danqi Deng
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, PR China
| | - Jianzhong Zhang
- Department of Dermatology, Peking University People's Hospital, Beijing, 100044, PR China
| |
Collapse
|
4
|
Kourkouni E, Tsiogkas SG, Mavropoulos A, Simopoulou T, Katsiari CG, Bogdanos DP, Sakkas LI. CD32 (FcγRIIB) expression is low on CD21 low B cells from systemic sclerosis patients with digital ulcers, interstitial lung disease, and anti-topoisomerase I autoantibodies. Clin Immunol 2024; 262:110195. [PMID: 38494058 DOI: 10.1016/j.clim.2024.110195] [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: 04/13/2023] [Revised: 03/11/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
CD21low B cells have recently been found increased in SSc-associated digital ulcers (DUs) or interstitial lung disease (ILD). To further characterize CD21low B cells which encompass autoreactive cells, we analyzed their expression of the inhibitory CD32 receptor in SSc. Peripheral blood mononuclear cells from 27 patients with SSc and 15 age-and sex-matched healthy controls (HCs) were analyzed with multicolor flow cytometry. CD21low B cells were significantly increased in patients with DUs (51.3%) compared to HCs (28.1%) and in patients with ILD (53.1%) compared to HCs. CD21lowCD32low B cells were significantly increased in patients with DUs (23.8%) compared to HCs (4.4%), in patients with ILD (28.4%) compared to HCs, and in anti-topoisomerase I (+) patients (21.5%) compared to HCs and to anti-topoisomerase I (-) patients (2.4%). Autoreactive B cells recognizing Topoisomerase I were predominantly within CD32low cell fraction. Our study further supports the autoreactive status of CD21lowCD32low B cells in SSc patients.
Collapse
Affiliation(s)
- Evangeli Kourkouni
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Sotirios G Tsiogkas
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Athanasios Mavropoulos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Theodora Simopoulou
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Christina G Katsiari
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Dimitrios P Bogdanos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Lazaros I Sakkas
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece; IASO Thessalias General Hospital, Larissa, Greece.
| |
Collapse
|
5
|
Merrill JT, Guthridge J, Smith M, June J, Koumpouras F, Machua W, Askanase A, Khosroshahi A, Sheikh SZ, Rathi G, Burington B, Foster P, Matijevic M, Arora S, Wang X, Gao M, Wax S, James JA, Zack DJ. Obexelimab in Systemic Lupus Erythematosus With Exploration of Response Based on Gene Pathway Co-Expression Patterns: A Double-Blind, Randomized, Placebo-Controlled, Phase 2 Trial. Arthritis Rheumatol 2023; 75:2185-2194. [PMID: 37459248 DOI: 10.1002/art.42652] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/17/2023] [Accepted: 07/11/2023] [Indexed: 11/30/2023]
Abstract
OBJECTIVE Obexelimab is an investigational, bifunctional, noncytolytic monoclonal antibody that binds CD19 and FcyRIIb to inhibit B cells, plasmablasts, and plasma cells. This trial evaluated the efficacy and safety of obexelimab in the treatment of patients with systemic lupus erythematosus (SLE). METHODS During screening, patients with active, non-organ-threatening SLE received corticosteroid injections to ameliorate symptoms while immunosuppressants were withdrawn (≤10 mg/day prednisone equivalent and ≤400 mg/day hydroxychloroquine allowed). Patients with improved disease activity were randomized 1:1 to obexelimab 5 mg/kg intravenously or placebo once every 2 weeks until week 32 or loss of improvement (LOI). RESULTS In this study, 104 patients were randomized. Analysis of the primary endpoint, proportion of patients reaching week 32 without LOI, used an efficacy-evaluable (EE) population defined as patients who completed the study or withdrew for flare or treatment-related toxicity. This endpoint did not reach statistical significance: 21 of 50 obexelimab-treated patients (42.0%) versus 12 of 42 patients (28.6%) treated with a placebo (P = 0.183). Time to LOI was increased in obexelimab-treated patients versus patients treated with a placebo in the EE (hazard ratio [HR] 0.53, P = 0.025) and intention-to-treat (HR 0.59, P = 0.062) populations. In obexelimab-treated patients, B cells decreased approximately 50%, and trough concentration and inclusion in baseline gene expression clusters with high B cell pathway modules were associated with increased time to LOI. Obexelimab was associated with infusion reactions but was generally safe and well-tolerated. CONCLUSION Although the primary endpoint was not reached, secondary analysis showed time to LOI was significantly increased in obexelimab-treated patients, and analysis of patient subsets defined by gene expression patterns at baseline suggests a responding subpopulation.
Collapse
Affiliation(s)
- Joan T Merrill
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Joel Guthridge
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Miles Smith
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Joshua June
- Great Lakes Center of Rheumatology, Lansing, Michigan
| | | | | | - Anca Askanase
- Columbia University Medical Center, New York City, New York
| | | | - Saira Z Sheikh
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | | | | | | | | | | | | | | | - Judith A James
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | | |
Collapse
|
6
|
Alemán OR, Rosales C. Human neutrophil Fc gamma receptors: different buttons for different responses. J Leukoc Biol 2023; 114:571-584. [PMID: 37437115 DOI: 10.1093/jleuko/qiad080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/31/2023] [Accepted: 06/28/2023] [Indexed: 07/14/2023] Open
Abstract
Neutrophils are fundamental cells in host defense. These leukocytes are quickly recruited from the blood to sites of infection or tissue damage. At these sites, neutrophils initiate several innate immune responses, including phagocytosis, production of reactive oxygen species, degranulation to release proteases and other antimicrobial compounds, production of inflammatory mediators, and formation of neutrophil extracellular traps. In addition to their role in innate immunity, neutrophils are now recognized as cells that also regulate adaptive immunity, via interaction with dendritic cells and lymphocytes. Neutrophils also respond to adaptive immunity by interacting with antibody molecules. Indeed, antibody molecules allow neutrophils to have antigen-specific responses. Neutrophils express different receptors for antibodies. The receptors for immunoglobulin G molecules are known as Fcγ receptors. Upon Fcγ receptor aggregation on the cell membrane, these receptors trigger distinct signal transduction cascades that activate particular cellular responses. In this review, we describe the major Fcγ receptors expressed on human neutrophils and discuss how each Fcγ receptor activates a choice of signaling pathways to stimulate particular neutrophil responses.
Collapse
Affiliation(s)
- Omar Rafael Alemán
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Apdo. Postal 70228, Ciudad Universitaria, Ciudad de México 04510, México
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Apdo. Postal 70228, Ciudad Universitaria, Ciudad de México 04510, México
| |
Collapse
|
7
|
Dadas O, Ertay A, Cragg MS. Delivering co-stimulatory tumor necrosis factor receptor agonism for cancer immunotherapy: past, current and future perspectives. Front Immunol 2023; 14:1147467. [PMID: 37180119 PMCID: PMC10167284 DOI: 10.3389/fimmu.2023.1147467] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/27/2023] [Indexed: 05/15/2023] Open
Abstract
The tumor necrosis factor superfamily (TNFSF) and their receptors (TNFRSF) are important regulators of the immune system, mediating proliferation, survival, differentiation, and function of immune cells. As a result, their targeting for immunotherapy is attractive, although to date, under-exploited. In this review we discuss the importance of co-stimulatory members of the TNFRSF in optimal immune response generation, the rationale behind targeting these receptors for immunotherapy, the success of targeting them in pre-clinical studies and the challenges in translating this success into the clinic. The efficacy and limitations of the currently available agents are discussed alongside the development of next generation immunostimulatory agents designed to overcome current issues, and capitalize on this receptor class to deliver potent, durable and safe drugs for patients.
Collapse
Affiliation(s)
- Osman Dadas
- Antibody and Vaccine Group, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ayse Ertay
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Mark S. Cragg
- Antibody and Vaccine Group, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
8
|
Kløve-Mogensen K, Steffensen R, Masmas TN, Glenthøj A, Jensen CF, Haunstrup TM, Ratcliffe P, Höglund P, Hasle H, Nielsen KR. Genetic variations in low-to-medium-affinity Fcγ receptors and autoimmune neutropenia in early childhood in a Danish cohort. Int J Immunogenet 2023; 50:65-74. [PMID: 36754570 DOI: 10.1111/iji.12614] [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: 11/25/2022] [Revised: 01/09/2023] [Accepted: 01/22/2023] [Indexed: 02/10/2023]
Abstract
Autoimmune neutropenia (AIN) in early childhood is caused by autoantibodies directed against antigens on the neutrophil membrane and is a frequent cause of neutropenia in children. Association of AIN with Fcγ receptor (FCGR) 3B variants is well described. In this study, we investigate genetic variations in the FCGR locus and copy number variation of FCGR3B. A total of 130 antibody-positive AIN patients, 64 with specific anti-HNA-1a antibodies and 66 with broad-reacting anti-FcγRIIIb antibodies, were genotyped with a multiplex ligation probe assay and compared with healthy controls. Positive findings were confirmed with real-time q-PCR. We determined copy numbers of the FCGR2 and FCGR3 genes and the following SNPs: FCGR2A Q62W (rs201218628), FCGR2A H166R (rs1801274), FCGR2B I232T (rs1050501), FCGR3A V176F (rs396991), haplotypes for FCGR2B/C promoters (rs3219018/rs780467580), FCGR2C STOP/ORF and HNA-1 genotypes in FCGR3B (rs447536, rs448740, rs52820103, rs428888 and rs2290834). Generally, associations were antibody specific, with all associations being representative of the anti-HNA-1a-positive group, while the only association found in the anti-FcγRIIIb group was with the HNA-1 genotype. An increased risk of AIN was observed for patients with one copy of FCGR3B; the HNA genotypes HNA-1a, HNA-1aa or HNA-1aac; the FCGR2A 166H and FCGR2B 232I variations; and no copies of FCGR2B 2B.4. A decreased risk was observed for HNA genotype HNA-1bb; FCGR2A 166R; FCGR2B 232T; and one copy of FCGR2B promoter 2B.4. We conclude that in our Danish cohort, there was a strong association between variation in the FCGR locus and AIN. The findings of different genetic associations between autoantibody groups could indicate the presence of two different disease entities and disease heterogeneity.
Collapse
Affiliation(s)
- Kirstine Kløve-Mogensen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Rudi Steffensen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Tania Nicole Masmas
- Pediatric Hematopoietic Stem Cell Transplantation and Immunodeficiency, Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Andreas Glenthøj
- Department of Hematology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Christina Friis Jensen
- Department of Pediatrics and Adolescent Medicine, Aalborg University Hospital, Aalborg, Denmark
| | - Thure Mors Haunstrup
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Paul Ratcliffe
- Department of medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Petter Höglund
- Department of medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Henrik Hasle
- Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Kaspar René Nielsen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
9
|
Zhuang T, Hu M, Wang J, Mei L, Zhu X, Zhang H, Jin F, Shao J, Wang T, Wang C, Niu X, Wu D. Sodium houttuyfonate effectively treats acute pulmonary infection of Pseudomonas aeruginosa by affecting immunity and intestinal flora in mice. Front Cell Infect Microbiol 2022; 12:1022511. [PMID: 36530439 PMCID: PMC9751016 DOI: 10.3389/fcimb.2022.1022511] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/14/2022] [Indexed: 12/04/2022] Open
Abstract
Introduction Pseudomonas aeruginosa is a major nosocomial pathogen that frequently causes ventilator-associated pneumonia in specific populations. Sodium houttuyfonate (SH) has shown mild antibacterial activity against P. aeruginosa in vitro, but the mechanism of potent antimicrobial activity of SH against P. aeruginosa infection in vivo remains unclear. Methods Here, using the mouse pneumonia model induced by P. aeruginosa nasal drip to explore the therapeutic effects of SH. Results We found that SH exhibits dose-dependent therapeutic effects of reducing P. aeruginosa burden and systemic inflammation in pneumonia mice. SH ameliorates inflammatory gene expression and production of inflammatory proteins, such as interleukin-6 (IL-6), nuclear factor kappa-B (NF-κB) and toll-like receptor 4 (TLR4), associated with the TLR4/NF-κB pathway in mice with P. aeruginosa pneumonia. Furthermore, we analyzed the intestinal flora of mice and found that compared with the model group, the abundance and diversity of beneficial bacterial flora of SH treatment groups increased significantly, suggesting that SH can improve the intestinal flora disorder caused by inflammation. In addition, SH improves alpha and beta diversity index and reduces species abundance differences of intestinal flora in pneumonia mice. Discussion Taken together, our presented results indicate that SH may effectively alleviate the acute pulmonary infection induced by P. aeruginosa by reducing the disturbance of regulating immunity and intestinal flora in mice.
Collapse
Affiliation(s)
- Tian Zhuang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China,Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China
| | - Mengxue Hu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China,Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China
| | - Jian Wang
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China,Pathology Department, First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Longfei Mei
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China,Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China
| | - Xiaoxiao Zhu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China,Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China
| | - Haitao Zhang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China,Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China
| | - Feng Jin
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China,Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China
| | - Jing Shao
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China,Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China,Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Tianming Wang
- Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Changzhong Wang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China,Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China,Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Xiaojia Niu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China,Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China,Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China,*Correspondence: Daqiang Wu, ; Xiaojia Niu,
| | - Daqiang Wu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China,Key Laboratory of Xin’an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China,*Correspondence: Daqiang Wu, ; Xiaojia Niu,
| |
Collapse
|
10
|
McLeish KR, Fernandes MJ. Understanding inhibitory receptor function in neutrophils through the lens of
CLEC12A. Immunol Rev 2022; 314:50-68. [PMID: 36424898 DOI: 10.1111/imr.13174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Neutrophils are the first leukocytes recruited from the circulation in response to invading pathogens or injured cells. To eradicate pathogens and contribute to tissue repair, recruited neutrophils generate and release a host of toxic chemicals that can also damage normal cells. To avoid collateral damage leading to tissue injury and organ dysfunction, molecular mechanisms evolved that tightly control neutrophil response threshold to activating signals, the strength and location of the response, and the timing of response termination. One mechanism of response control is interruption of activating intracellular signaling pathways by the 20 inhibitory receptors expressed by neutrophils. The two inhibitory C-type lectin receptors expressed by neutrophils, CLEC12A and DCIR, exhibit both common and distinct molecular and functional mechanisms, and they are associated with different diseases. In this review, we use studies on CLEC12A as a model of inhibitory receptor regulation of neutrophil function and participation in disease. Understanding the molecular mechanisms leading to inhibitory receptor specificity offers the possibility of using physiologic control of neutrophil functions as a pharmacologic tool to control inflammatory diseases.
Collapse
Affiliation(s)
- Kenneth R. McLeish
- Department of Medicine University of Louisville School of Medicine Louisville Kentucky USA
| | - Maria J. Fernandes
- Infectious and Immune Diseases Division CHU de Québec‐Laval University Research Center Québec Québec Canada
- Department of Microbiology‐Infectious Diseases and Immunology, Faculty of Medicine Laval University Québec Québec Canada
| |
Collapse
|
11
|
Petty A, Glass LJ, Rothmond DA, Purves-Tyson T, Sweeney A, Kondo Y, Kubo S, Matsumoto M, Weickert CS. Increased levels of a pro-inflammatory IgG receptor in the midbrain of people with schizophrenia. J Neuroinflammation 2022; 19:188. [PMID: 35841099 PMCID: PMC9287858 DOI: 10.1186/s12974-022-02541-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND There is growing evidence that neuroinflammation may contribute to schizophrenia neuropathology. Elevated pro-inflammatory cytokines are evident in the midbrain from schizophrenia subjects, findings that are driven by a subgroup of patients, characterised as a "high inflammation" biotype. Cytokines trigger the release of antibodies, of which immunoglobulin G (IgG) is the most common. The level and function of IgG is regulated by its transporter (FcGRT) and by pro-inflammatory IgG receptors (including FcGR3A) in balance with the anti-inflammatory IgG receptor FcGR2B. Testing whether abnormalities in IgG activity contribute to the neuroinflammatory abnormalities schizophrenia patients, particularly those with elevated cytokines, may help identify novel treatment targets. METHODS Post-mortem midbrain tissue from healthy controls and schizophrenia cases (n = 58 total) was used to determine the localisation and abundance of IgG and IgG transporters and receptors in the midbrain of healthy controls and schizophrenia patients. Protein levels of IgG and FcGRT were quantified using western blot, and gene transcript levels of FcGRT, FcGR3A and FcGR2B were assessed using qPCR. The distribution of IgG in the midbrain was assessed using immunohistochemistry and immunofluorescence. Results were compared between diagnostic (schizophrenia vs control) and inflammatory (high vs low inflammation) groups. RESULTS We found that IgG and FcGRT protein abundance (relative to β-actin) was unchanged in people with schizophrenia compared with controls irrespective of inflammatory subtype. In contrast, FcGRT and FcGR3A mRNA levels were elevated in the midbrain from "high inflammation" schizophrenia cases (FcGRT; p = 0.02, FcGR3A; p < 0.0001) in comparison to low-inflammation patients and healthy controls, while FcGR2B mRNA levels were unchanged. IgG immunoreactivity was evident in the midbrain, and approximately 24% of all individuals (control subjects and schizophrenia cases) showed diffusion of IgG from blood vessels into the brain. However, the intensity and distribution of IgG was comparable across schizophrenia cases and control subjects. CONCLUSION These findings suggest that an increase in the pro-inflammatory Fcγ receptor FcGR3A, rather than an overall increase in IgG levels, contribute to midbrain neuroinflammation in schizophrenia patients. However, more precise information about IgG-Fcγ receptor interactions is needed to determine their potential role in schizophrenia neuropathology.
Collapse
Affiliation(s)
- A Petty
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, NSW, 2031, Australia
- School of Psychiatry, University of New South Wales, Sydney, NSW, 2052, Australia
| | - L J Glass
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, NSW, 2031, Australia
- Centre for Immunology and Allergy Research, Westmead Institute of Medical Research, The University of Sydney, Sydney, Australia
| | - D A Rothmond
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, NSW, 2031, Australia
| | - T Purves-Tyson
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, NSW, 2031, Australia
- School of Psychiatry, University of New South Wales, Sydney, NSW, 2052, Australia
| | - A Sweeney
- NSW Brain Tissue Resource Centre, University of Sydney, Sydney, NSW, 2006, Australia
| | - Y Kondo
- Astellas Research Institute of America LLC, San Diego, CA, 92121, USA
| | - S Kubo
- Astellas Pharma Inc., Tsukuba, Ibaraki, 305-8585, Japan
| | - M Matsumoto
- Astellas Research Institute of America LLC, San Diego, CA, 92121, USA
| | - C Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Sydney, NSW, 2031, Australia.
- School of Psychiatry, University of New South Wales, Sydney, NSW, 2052, Australia.
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
12
|
Barlev AN, Malkiel S, Kurata-Sato I, Dorjée AL, Suurmond J, Diamond B. FcɣRIIB regulates autoantibody responses by limiting marginal zone B cell activation. J Clin Invest 2022; 132:157250. [PMID: 35819855 PMCID: PMC9435648 DOI: 10.1172/jci157250] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
FcɣRIIB is an inhibitory receptor expressed throughout B cell development. Diminished expression or function is associated with lupus in mice and humans, in particular through an effect on autoantibody production and plasma cell differentiation. Here, we analysed the effect of B cell-intrinsic FcɣRIIB expression on B cell activation and plasma cell differentiation. Loss of FcɣRIIB on B cells (Fcgr2b cKO mice) led to a spontaneous increase in autoantibody titers. This increase was most striking for IgG3, suggestive of increased extrafollicular responses. Marginal zone (MZ) B cells had the highest expression of FcɣRIIB in both mouse and human. This high expression of FcɣRIIB was linked to increased MZ B cell activation, Erk phosphorylation, and calcium fluxin the absence of FcɣRIIB triggering. Marked increases in IgG3+ plasma cells and B cells were observed during extrafollicular plasma cell responses in Fcgr2b cKO mice. The increased IgG3 response following immunization of Fcgr2b cKO mice was lost in MZ-deficient Notch2/Fcgr2b cKO mice. Importantly, SLE patients exhibited decreased expression of FcɣRIIB, most strongly in MZ B cells. Thus, we present a model where high FcɣRIIB expression in MZ B cells prevents their hyperactivation and ensuing autoimmunity.
Collapse
Affiliation(s)
- Ashley N Barlev
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Susan Malkiel
- Center of Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, United States of America
| | - Izumi Kurata-Sato
- Center of Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, United States of America
| | - Annemarie L Dorjée
- Department of Rheumatology, Leiden University Medical Center, Leiden, Netherlands
| | - Jolien Suurmond
- Department of Rheumatology, Leiden University Medical Center, Leiden, Netherlands
| | - Betty Diamond
- Center of Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, United States of America
| |
Collapse
|
13
|
Baecher KM, Ford ML. Intersection of FcγRIIB, the microbiome, and checkpoint inhibitors in antitumor immunity. Cancer Immunol Immunother 2021; 70:3397-3404. [PMID: 34241677 PMCID: PMC10992943 DOI: 10.1007/s00262-021-03004-4] [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: 09/01/2020] [Accepted: 06/29/2021] [Indexed: 10/20/2022]
Abstract
Fc receptors (FcRs) and the microbiome are both known to have an effect on the development and progression of cancers. Checkpoint inhibitors are a novel class of therapeutics which are used to combat cancer and are integrally linked to both FcRs and the microbiome. The use of checkpoint inhibitors has grown exponentially over the past decade, although many host factors affect both the efficacy and the safety of these therapeutics. Some of these host factors, including the microbiome and the expression of FcRs, are currently being investigated. Here we discuss the current understanding of FcRs (particularly the inhibitory FcγRIIB) and the microbiome in context of T cell immunity, inflammation, cancer, and checkpoint inhibition.
Collapse
Affiliation(s)
- Kirsten M Baecher
- Division of Transplant, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Mandy L Ford
- Division of Transplant, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
14
|
Wolf B, Jeliazkova-Mecheva V, Del Rio-Espinola A, Boisclair J, Walker D, Cochin De Billy B, Flaherty M, Flandre T. An afucosylated anti-CD32b monoclonal antibody induced platelet-mediated adverse events in a human Fcγ receptor transgenic mouse model and its potential human translatability. Toxicol Sci 2021; 185:89-104. [PMID: 34687301 DOI: 10.1093/toxsci/kfab124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
To assess the safety and tolerability of NVS32b, a monoclonal, afucosylated, anti-CD32b (FCGR2B) antibody we used a humanized transgenic (Tg) mouse model that expresses all human Fc gamma receptors (FCGRs) while lacking all mouse FCGRs. Prior to its use, we extensively characterized the model. We found expression of all human FCGRs in a pattern similar to humans with some exceptions, such as low CD32 expression on T cells (detected with the pan CD32 antibody but more notably with the CD32b-specific antibody), variation in the transgene copy number, integration of additional human genes, and overall higher expression of all FCGRs on myeloid cells compared to human. Unexpectedly, NVS32b induced severe acute generalized thrombosis in huFCGR mice upon iv dosing. Mechanistic evaluation on huFCGR and human platelets revealed distinct binding, activation and aggregation driven by NVS32b in both species. In huFCGR mice, the anti-CD32b antibody NVS32b binds platelet CD32a via both Fc and/or CDR (complementarity determining region) causing their activation while in human, NVS32b-binding requires platelet pre-activation and interaction of platelet CD32a via the Fc portion and an unknown platelet epitope via the CDR portion of NVS32b. We deemed the huFCGR mice to be over-predictive of the NVS32b-associated human thrombotic risk. Impact: In this study we elucidated the mechanism based on the thrombotic adverse events observed in huFCGR mice upon NVS32B dosing and were able to identify this safety liability which led to program termination. Therefore, this mouse model could be useful in research of immunotherapies targeting or involving FCGRs. Potential biological implications resulting from species differences in the FCGR expression pattern are nevertheless important to consider.
Collapse
Affiliation(s)
- B Wolf
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | | | - J Boisclair
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - D Walker
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | | | - M Flaherty
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - T Flandre
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| |
Collapse
|
15
|
Macri C, Morgan H, Villadangos JA, Mintern JD. Regulation of dendritic cell function by Fc-γ-receptors and the neonatal Fc receptor. Mol Immunol 2021; 139:193-201. [PMID: 34560415 DOI: 10.1016/j.molimm.2021.07.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 06/28/2021] [Accepted: 07/19/2021] [Indexed: 01/02/2023]
Abstract
Dendritic cells (DCs) express receptors to sense pathogens and/or tissue damage and to communicate with other immune cells. Among those receptors, Fc receptors (FcRs) are triggered by the Fc region of antibodies produced during adaptive immunity. In this review, the role of FcγR and neonatal Fc receptor (FcRn) in DC immunity will be discussed. Their expression in DC subsets and impact on antigen uptake and presentation, DC maturation and polarisation of T cell responses will be described. Lastly, we will discuss the importance of FcR-mediated DC function in the context of immunity during viral infection, inflammatory disease, cancer and immunotherapy.
Collapse
Affiliation(s)
- Christophe Macri
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia
| | - Huw Morgan
- ACRF Translational Research Laboratory, The Royal Melbourne Hospital, Parkville, Melbourne, Victoria, 3050, Australia; Department of Medicine, University of Melbourne, Parkville, Melbourne, Victoria, 3010, Australia
| | - Jose A Villadangos
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia; Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, 3010, Australia.
| | - Justine D Mintern
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
16
|
Identification of candidate regulators of mandibular bone loss in FcγRIIB -/- Mice. Sci Rep 2021; 11:18726. [PMID: 34548536 PMCID: PMC8455620 DOI: 10.1038/s41598-021-98108-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/25/2021] [Indexed: 11/15/2022] Open
Abstract
Patients with systemic lupus erythematosus (SLE) have increased inflammatory cytokines, leading to periodontitis and alveolar bone loss. However, the mechanisms driving this phenomenon are still unknown. Here, we have identified novel therapeutic targets for and mediators of lupus-mediated bone loss using RNA-sequencing (RNA-seq) in a FcγRIIB-/- mouse model of lupus associated osteopenia. A total of 2,710 upregulated and 3,252 downregulated DEGs were identified. The GO and KEGG annotations revealed that osteoclast differentiation, bone mineralization, ossification, and myeloid cell development were downregulated. WikiPathways indicated that Hedgehog, TNFα NF-κB and Notch signaling pathway were also decreased. We identified downregulated targets, Sufu and Serpina12, that have important roles in bone homeostasis. Sufu and Serpina12 were related to Hedgehog signaling proteins, including Gli1, Gli2, Gli3, Ptch1, and Ptch2. Gene knockdown analysis demonstrated that Sufu, and Serpina12 contributed to osteoclastogenesis and osteoblastogenesis, respectively. Osteoclast and osteoblast marker genes were significantly decreased in Sufu-deficient and Serpina12-deficient cells, respectively. Our results suggest that alterations in Hedgehog signaling play an important role in the pathogenesis of osteopenia in FcγRIIB-/- mice. The novel DEGs and pathways identified in this study provide new insight into the underlying mechanisms of mandibular bone loss during lupus development.
Collapse
|
17
|
Dörner T, Szelinski F, Lino AC, Lipsky PE. Therapeutic implications of the anergic/postactivated status of B cells in systemic lupus erythematosus. RMD Open 2021; 6:rmdopen-2020-001258. [PMID: 32675278 PMCID: PMC7425190 DOI: 10.1136/rmdopen-2020-001258] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/28/2020] [Accepted: 06/12/2020] [Indexed: 12/19/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is characterised by numerous abnormalities in B lineage cells, including increased CD27++ plasmablasts/plasma cells, atypical CD27-IgD- B cells with increased CD95, spleen tyrosine kinase (Syk)++, CXCR5- and CXCR5+ subsets and anergic CD11c+Tbet+ age-associated B cells. Most findings, together with preclinical lupus models, support the concept of B cell hyperactivity in SLE. However, it remains largely unknown whether these specific B cell subsets have pathogenic consequences and whether they provide relevant therapeutic targets. Recent findings indicate a global distortion of B cell functional capability, in which the entire repertoire of naïve and memory B cells in SLE exhibits an anergic or postactivated (APA) functional phenotype. The APA status of SLE B cells has some similarities to the functional derangement of lupus T cells. APA B cells are characterised by reduced global cytokine production, diminished B cell receptor (BCR) signalling with decreased Syk and Bruton's tyrosine kinase phosphorylation related to repeated in vivo BCR stimulation as well as hyporesponsiveness to toll-like receptor 9 engagement, but intact CD40 signalling. This APA status was related to constitutive co-localisation of CD22 linked to phosphatase SHP-1 and increased overall protein phosphatase activities. Notably, CD40 co-stimulation could revert this APA status and restore BCR signalling, downregulate protein tyrosine phosphatase transcription and promote B cell proliferation and differentiation. The APA status and their potential rescue by bystander help conveyed through CD40 stimulation not only provides insights into possible mechanisms of escape of autoreactive clones from negative selection but also into novel ways to target B cells therapeutically.
Collapse
Affiliation(s)
| | | | - Andreia C Lino
- Department of Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany.,German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Peter E Lipsky
- RILITE Research Institute, Charlottesville, Virginia, USA
| |
Collapse
|
18
|
Turman JM, Cheplowitz AM, Tiwari C, Thomas T, Joshi D, Bhat M, Wu Q, Pong E, Chu SY, Szymkowski DE, Sharma A, Seveau S, Robinson JM, Kwiek JJ, Burton D, Rajaram MVS, Kim J, Hangartner L, Ganesan LP. Accelerated Clearance and Degradation of Cell-Free HIV by Neutralizing Antibodies Occurs via FcγRIIb on Liver Sinusoidal Endothelial Cells by Endocytosis. THE JOURNAL OF IMMUNOLOGY 2021; 206:1284-1296. [PMID: 33568400 DOI: 10.4049/jimmunol.2000772] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 01/05/2021] [Indexed: 01/19/2023]
Abstract
Neutralizing Abs suppress HIV infection by accelerating viral clearance from blood circulation in addition to neutralization. The elimination mechanism is largely unknown. We determined that human liver sinusoidal endothelial cells (LSEC) express FcγRIIb as the lone Fcγ receptor, and using humanized FcγRIIb mouse, we found that Ab-opsonized HIV pseudoviruses were cleared considerably faster from circulation than HIV by LSEC FcγRIIb. Compared with humanized FcγRIIb-expressing mice, HIV clearance was significantly slower in FcγRIIb knockout mice. Interestingly, a pentamix of neutralizing Abs cleared HIV faster compared with hyperimmune anti-HIV Ig (HIVIG), although the HIV Ab/Ag ratio was higher in immune complexes made of HIVIG and HIV than pentamix and HIV. The effector mechanism of LSEC FcγRIIb was identified to be endocytosis. Once endocytosed, both Ab-opsonized HIV pseudoviruses and HIV localized to lysosomes. This suggests that clearance of HIV, endocytosis, and lysosomal trafficking within LSEC occur sequentially and that the clearance rate may influence downstream events. Most importantly, we have identified LSEC FcγRIIb-mediated endocytosis to be the Fc effector mechanism to eliminate cell-free HIV by Abs, which could inform development of HIV vaccine and Ab therapy.
Collapse
Affiliation(s)
- James M Turman
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Alana M Cheplowitz
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Charu Tiwari
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Thushara Thomas
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Dhruvi Joshi
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Menakshi Bhat
- Center for Retrovirus Research, Department of Microbiology, The Ohio State University, Columbus, OH 43210
| | - Qian Wu
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210
| | | | | | | | - Amit Sharma
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210.,Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Stephanie Seveau
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210
| | - John M Robinson
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH 43210
| | - Jesse J Kwiek
- Center for Retrovirus Research, Department of Microbiology, The Ohio State University, Columbus, OH 43210
| | - Dennis Burton
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037; and
| | - Murugesan V S Rajaram
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210
| | - Jonghan Kim
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115
| | - Lars Hangartner
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037; and
| | - Latha P Ganesan
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210;
| |
Collapse
|
19
|
Bhamidipati K, Silberstein JL, Chaichian Y, Baker MC, Lanz TV, Zia A, Rasheed YS, Cochran JR, Robinson WH. CD52 Is Elevated on B cells of SLE Patients and Regulates B Cell Function. Front Immunol 2021; 11:626820. [PMID: 33658999 PMCID: PMC7917337 DOI: 10.3389/fimmu.2020.626820] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/17/2020] [Indexed: 11/13/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by B cell dysregulation and breaks in tolerance that lead to the production of pathogenic autoantibodies. We performed single-cell RNA sequencing of B cells from healthy donors and individuals with SLE which revealed upregulated CD52 expression in SLE patients. We further demonstrate that SLE patients exhibit significantly increased levels of B cell surface CD52 expression and plasma soluble CD52, and levels of soluble CD52 positively correlate with measures of lupus disease activity. Using CD52-deficient JeKo-1 cells, we show that cells lacking surface CD52 expression are hyperresponsive to B cell receptor (BCR) signaling, suggesting an inhibitory role for the surface-bound protein. In healthy donor B cells, antigen-specific BCR-activation initiated CD52 cleavage in a phospholipase C dependent manner, significantly reducing cell surface levels. Experiments with recombinant CD52-Fc showed that soluble CD52 inhibits BCR signaling in a manner partially-dependent on Siglec-10. Moreover, incubation of unstimulated B cells with CD52-Fc resulted in the reduction of surface immunoglobulin and CXCR5. Prolonged incubation of B cells with CD52 resulted in the expansion of IgD+IgMlo anergic B cells. In summary, our findings suggest that CD52 functions as a homeostatic protein on B cells, by inhibiting responses to BCR signaling. Further, our data demonstrate that CD52 is cleaved from the B cell surface upon antigen engagement, and can suppress B cell function in an autocrine and paracrine manner. We propose that increased expression of CD52 by B cells in SLE represents a homeostatic mechanism to suppress B cell hyperactivity.
Collapse
Affiliation(s)
- Kartik Bhamidipati
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, United States
- VA Palo Alto Healthcare System, Palo Alto, CA, United States
- Division of Immunology and Rheumatology, School of Medicine, Stanford University, Stanford, CA, United States
| | - John L. Silberstein
- Program in Immunology, Stanford University School of Medicine, Stanford, CA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Yashaar Chaichian
- Division of Immunology and Rheumatology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Matthew C. Baker
- Division of Immunology and Rheumatology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Tobias V. Lanz
- VA Palo Alto Healthcare System, Palo Alto, CA, United States
- Division of Immunology and Rheumatology, School of Medicine, Stanford University, Stanford, CA, United States
- Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Amin Zia
- VA Palo Alto Healthcare System, Palo Alto, CA, United States
- Division of Immunology and Rheumatology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Yusuf S. Rasheed
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Jennifer R. Cochran
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - William H. Robinson
- VA Palo Alto Healthcare System, Palo Alto, CA, United States
- Division of Immunology and Rheumatology, School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
20
|
B Cell Aberrance in Lupus: the Ringleader and the Solution. Clin Rev Allergy Immunol 2021; 62:301-323. [PMID: 33534064 DOI: 10.1007/s12016-020-08820-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2020] [Indexed: 12/18/2022]
Abstract
Systemic lupus erythematosus (SLE) is a prototypical autoimmune disease with high heterogeneity but the common characterization of numerous autoantibodies and systemic inflammation which lead to the damage of multiple organs. Aberrance of B cells plays a pivotal role in the immunopathogenesis of SLE via both antibody-dependent and antibody-independent manners. Escape of autoreactive B cells from the central and peripheral tolerance checkpoints, over-activation of B cells and their excessive cytokines release which drive T cells and dendritic cells stimulation, and dysregulated surface molecules, as well as intracellular signal pathways involved in B cell biology, are all contributing to B cell aberrance and participating in the pathogenesis of SLE. Based on that rationale, targeting aberrance of B cells and relevant molecules and pathways is expected to be a promising strategy for lupus control. Multiple approaches targeting B cells through different mechanisms have been attempted, including B-cell depletion via monoclonal antibodies against B-cell-specific molecules, blockade of B-cell survival and activation factors, suppressing T-B crosstalk by interrupting costimulatory molecules and inhibiting intracellular activation signaling cascade by targeting pathway molecules in B cells. Though most attempts ended in failure, the efficacy of B-cell targeting has been encouraged by the FDA approval of belimumab that blocks B cell-activating factor (BAFF) and the recommended use of anti-CD20 as a remedial therapy in refractory lupus. Still, quantities of clinical trials targeting B cells or relevant molecules are ongoing and some of them have displayed promising preliminary results. Additionally, advances in multi-omics studies help deepen our understandings of B cell biology in lupus and may promote the discovery of novel potential therapeutic targets. The combination of real-world data with basic research achievements may pave the road to conquering lupus.
Collapse
|
21
|
Chu SY, Pong E, Bonzon C, Yu N, Jacob CO, Chalmers SA, Putterman C, Szymkowski DE, Stohl W. Inhibition of B cell activation following in vivo co-engagement of B cell antigen receptor and Fcγ receptor IIb in non-autoimmune-prone and SLE-prone mice. J Transl Autoimmun 2021; 4:100075. [PMID: 33409482 PMCID: PMC7773957 DOI: 10.1016/j.jtauto.2020.100075] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/09/2020] [Indexed: 11/27/2022] Open
Abstract
Engagement of Fcγ receptor IIb (FcγRIIb) suppresses B cell activation and represents a promising target for therapy in autoimmunity. Obexelimab is a non-depleting anti-human CD19 mAb with an Fc region engineered to have high affinity for human FcγRIIb, thereby co-engaging BCR and FcγRIIb. To assess its ability to suppress B cell activation in vivo, we generated non-autoimmune-prone C57BL/6 (B6) and SLE-prone NZM 2328 (NZM) mice in which the human FcγRIIb extracellular domain was knocked into the mouse Fcgr2b locus (B6.hRIIb and NZM.hRIIb mice, respectively, the latter retaining features of SLE). XENP8206, a mAb which bears the same FcγRIIb-enhanced human Fc domain as does obexelimab but which recognizes murine CD19 rather than human CD19, inhibited in vitro BCR-triggered activation of B cells from both B6.hRIIb and NZM.hRIIb mice. Following administration of XENP8206 to B6.hRIIb or NZM.hRIIb mice, B cell numbers in the spleen and lymph nodes remained stable but became hyporesponsive to BCR-triggered activation for at least 14 days. These findings demonstrate proof-of-principle that pharmacologic co-engagement of BCR and human FcγRIIb inhibits B cell activation in non-autoimmune and SLE-prone hosts while preserving B cell numbers. These observations lay a strong foundation for clinical trials in human SLE with agents that co-engage BCR and FcγRIIb. Moreover, B6.hRIIb and NZM.hRIIb should serve as powerful in vivo models in the elucidation of the cellular and molecular underpinnings of the changes induced by BCR/FcγRIIb co-engagement. We generated non-autoimmune B6.hRIIb and SLE-prone NZM.hRIIb knockin mice for the human FcγRIIb extracellular domain. XENP8206 is an anti-murine CD19 mAb engineered to have high affinity for human FcγRIIb. XENP8206 inhibited in vitro BCR-triggered activation of B cells from both B6.hRIIb and NZM.hRIIb mice. XENP8206 inhibited in vivo BCR-triggered activation of B cells while preserving B cell numbers. These observations lay a strong foundation for clinical trials in human SLE with agents that co-engage BCR and FcγRIIb.
Collapse
Affiliation(s)
| | - Erik Pong
- Xencor, Inc., Monrovia, CA, 91016, USA
| | | | - Ning Yu
- Division of Rheumatology, Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Chaim O Jacob
- Division of Rheumatology, Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Samantha A Chalmers
- Division of Rheumatology and Department of Microbiology and Immunology, Albert Einstein School of Medicine, Bronx, NY, 10461, USA
| | - Chaim Putterman
- Division of Rheumatology and Department of Microbiology and Immunology, Albert Einstein School of Medicine, Bronx, NY, 10461, USA
| | | | - William Stohl
- Division of Rheumatology, Department of Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| |
Collapse
|
22
|
Wang H, Teng X, Abboud G, Li W, Ye S, Morel L. D-mannose ameliorates autoimmune phenotypes in mouse models of lupus. BMC Immunol 2021; 22:1. [PMID: 33402096 PMCID: PMC7786459 DOI: 10.1186/s12865-020-00392-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/27/2020] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Systemic lupus erythematosus is an autoimmune disease characterized by an overproduction of autoantibodies resulting from dysregulation in multiple immune cell types. D-mannose is a C- 2 epimer of glucose that exhibits immunoregulatory effects in models of autoimmune diseases, such as type 1 diabetes, induced rheumatoid arthritis, and airway inflammation. This study was conducted to evaluate the efficacy of D-mannose treatment in mouse models of lupus. RESULTS Firstly, the effect of D-Mannose was evaluated by flow cytometry on the in vitro activation of non-autoimmune C57BL/6 (B6) bone marrow-derived dendritic cells (BMDCs) and their ability to induce antigen-specific CD4+ T cell proliferation and activation. D-mannose inhibited the maturation of BMDCs and their induction of antigen-specific T cell proliferation and activation. In vivo, D-mannose increased the frequency of Foxp3+ regulatory T cells in unmanipulated B6 mice. To assess the effect of D-mannose in mouse models of lupus, we used the graft-versus-host disease (cGVHD) induced model and the B6.lpr spontaneous model. In the cGVHD model, D-mannose treatment decreased autoantibody production, with a concomitant reduction of the frequency of effector memory and follicular helper T cells as well as germinal center B cells and plasma cells. These results were partially validated in the B6.lpr model of spontaneous lupus. CONCLUSION Overall, our results suggest that D-mannose ameliorates autoimmune activation in models of lupus, at least partially due to its expansion of Treg cells, the induction of immature conventional dendritic cells and the downregulation of effector T cells activation. D-Mannose showed however a weaker immunomodulatory effect in lupus than in other autoimmune diseases.
Collapse
Affiliation(s)
- Haiting Wang
- Department of Rheumatology, RenJi Hospital South Campus, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiangyu Teng
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, JHMHC 275, Box 100275, Gainesville, FL, 32610-0275, USA
| | - Georges Abboud
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, JHMHC 275, Box 100275, Gainesville, FL, 32610-0275, USA
| | - Wei Li
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, JHMHC 275, Box 100275, Gainesville, FL, 32610-0275, USA
| | - Shuang Ye
- Department of Rheumatology, RenJi Hospital South Campus, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Laurence Morel
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, JHMHC 275, Box 100275, Gainesville, FL, 32610-0275, USA.
| |
Collapse
|
23
|
Amiah MA, Ouattara A, Okou DT, N'Guetta SPA, Yavo W. Polymorphisms in Fc Gamma Receptors and Susceptibility to Malaria in an Endemic Population. Front Immunol 2020; 11:561142. [PMID: 33281811 PMCID: PMC7689034 DOI: 10.3389/fimmu.2020.561142] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/05/2020] [Indexed: 11/13/2022] Open
Abstract
Repeated infections by Plasmodium falciparum result in a humoral response that could reduce disease symptoms and prevent the development of clinical malaria. The principal mechanism underlying this humoral response is that immunoglobulin G (IgG) binds directly to the parasites, thus causing their neutralization. However, the action of antibodies alone is not always sufficient to eliminate pathogens from an organism. One key element involved in the recognition of IgG that plays a crucial role in the destruction of the parasites responsible for spreading malaria is the family of Fc gamma receptors. These receptors are expressed on the surface of immune cells. Several polymorphisms have been detected in the genes encoding these receptors, associated with susceptibility or resistance to malaria in different populations. In this review, we describe identified polymorphisms within the family of Fc gamma receptors and the impact of these variations on the response of a host to infection as well as provide new perspectives for the design of an effective vaccine for malaria.
Collapse
Affiliation(s)
- Mireille Ahou Amiah
- Malaria Research and Control Center, National Public Health Institute, Abidjan, Côte d'Ivoire.,Laboratory of Genetics, Unité de Formation et de Recherche (UFR) BIOSCIENCES, Félix Houphouët-Boigny University, Abidjan, Côte d'Ivoire
| | - Amed Ouattara
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - David Tea Okou
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Simon-Pierre Assanvo N'Guetta
- Laboratory of Genetics, Unité de Formation et de Recherche (UFR) BIOSCIENCES, Félix Houphouët-Boigny University, Abidjan, Côte d'Ivoire
| | - William Yavo
- Malaria Research and Control Center, National Public Health Institute, Abidjan, Côte d'Ivoire.,Department of Parasitology and Mycology, Faculty of Pharmacy, Félix Houphouët-Boigny University, Abidjan, Côte d'Ivoire
| |
Collapse
|
24
|
Monoclonal Antibody to CD14, TLR4, or CD11b: Impact of Epitope and Isotype Specificity on ROS Generation by Human Granulocytes and Monocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5708692. [PMID: 33294123 PMCID: PMC7700042 DOI: 10.1155/2020/5708692] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/24/2020] [Accepted: 11/10/2020] [Indexed: 02/01/2023]
Abstract
Lipopolysaccharides (LPSs or endotoxins) from Gram-negative bacteria represent pathogen-associated molecular patterns (PAMPs) that are recognized by CD14 and Toll-like receptor 4 (TLR4). Lipopolysaccharides prime polymorphonuclear leukocytes (PMNs) for substantial production of reactive oxygen species (ROS) during its response to secondary stimuli such as chemoattractants or pathogens. The excessive ROS production can damage surrounding host tissues, thereby amplifying the inflammatory reaction caused by pathogens. Today, specific antibodies against CD14, TLR4, and CD11b are being used as the essential tools to elucidate the role of these receptors in acute inflammation and some of these antibodies have advised as therapeutic agents for clinical use. Because each antibody has two antigen-binding arms [F(ab′)2] and one Fc arm, its effect on cellular response is much more complicated rather than simple blockage of target receptor. In fact, IgG antibody, once bound to target receptor, engages Fc receptors γ (FcγRs) and thereby is able to activate the adaptive immune system. The consequences of antibody-dependent binary heterotypic association of CD14, TLR4, or CD11b with FcγRs as well as homotypic one on ROS production are not well elucidated. Moreover, the consequences of antigenic recognition of CD14, TLR4, or CD11b by specific F(ab′)2 fragments are not always investigated. In this review, we will discuss known mechanisms underlying the therapeutic efficiency of CD14, TLR4, and CD11b/CD18 antibodies with a focus on LPS-dependent ROS or cytokine production by PMNs or monocytes. The impacts of F(ab′)2 as well as antibody IgG subclasses (isotypes) in therapeutic efficiency or agonistic potency of known antibodies against abovementioned receptors are presented. We also pay attention to how the efficiency of different IgG antibody subclasses is modulated during LPS-induced inflammation and by production of priming agents such as interferon γ (IFN-γ). Our review reinforces the molecular targets and therapeutic approaches to amelioration of harmful consequences of excessive activation of human pattern recognition receptors.
Collapse
|
25
|
Ustyanovska Avtenyuk N, Visser N, Bremer E, Wiersma VR. The Neutrophil: The Underdog That Packs a Punch in the Fight against Cancer. Int J Mol Sci 2020; 21:E7820. [PMID: 33105656 PMCID: PMC7659937 DOI: 10.3390/ijms21217820] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/19/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
The advent of immunotherapy has had a major impact on the outcome and overall survival in many types of cancer. Current immunotherapeutic strategies typically aim to (re)activate anticancer T cell immunity, although the targeting of macrophage-mediated anticancer innate immunity has also emerged in recent years. Neutrophils, although comprising ≈ 60% of all white blood cells in the circulation, are still largely overlooked in this respect. Nevertheless, neutrophils have evident anticancer activity and can induce phagocytosis, trogocytosis, as well as the direct cytotoxic elimination of cancer cells. Furthermore, therapeutic tumor-targeting monoclonal antibodies trigger anticancer immune responses through all innate Fc-receptor expressing cells, including neutrophils. Indeed, the depletion of neutrophils strongly reduced the efficacy of monoclonal antibody treatment and increased tumor progression in various preclinical studies. In addition, the infusion of neutrophils in murine cancer models reduced tumor progression. However, evidence on the anticancer effects of neutrophils is fragmentary and mostly obtained in in vitro assays or murine models with reports on anticancer neutrophil activity in humans lagging behind. In this review, we aim to give an overview of the available knowledge of anticancer activity by neutrophils. Furthermore, we will describe strategies being explored for the therapeutic activation of anticancer neutrophil activity.
Collapse
Affiliation(s)
| | | | - Edwin Bremer
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen (UMCG), University of Groningen, Hanzeplein 1/DA13, 9713 GZ Groningen, The Netherlands; (N.U.A.); (N.V.)
| | - Valerie R. Wiersma
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen (UMCG), University of Groningen, Hanzeplein 1/DA13, 9713 GZ Groningen, The Netherlands; (N.U.A.); (N.V.)
| |
Collapse
|
26
|
Luo S, Long H, Lu Q. Recent advances in understanding pathogenesis and therapeutic strategies of Systemic Lupus Erythematosus. Int Immunopharmacol 2020; 89:107028. [PMID: 33039962 DOI: 10.1016/j.intimp.2020.107028] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/30/2020] [Accepted: 09/17/2020] [Indexed: 01/07/2023]
Abstract
Systemic lupus erythematosus (SLE) is a multi-system-involving autoimmune disorder mainly affecting young and middle-aged women. Autoantibodies formation and immune complex deposition as well as other immune mechanisms contribute to heterogeneous clinical presentation, which leads to challenges for diagnosis and management. Although the exact pathogenesis of SLE is highly complicated, the pathophysiological understanding of SLE is constantly evolving and relevant studies were continually published, providing a better understanding of the molecular mechanisms. Moreover, new therapeutic strategies and management plans targeting pivotal factors involved in the pathogenesis of SLE got well established recently. In this article, we reviewed recent studies to provide an update in understanding pathogenesis and therapeutic strategies of SLE.
Collapse
Affiliation(s)
- Shuaihantian Luo
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Hai Long
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
27
|
Huang L, Yu X, Li L, Liu J, Wu X, Zeng Y, Liao X, Liu W, Zhang F, Zhang X, Chen H, Zheng W. Aberrant FcγRIIb and FcγRIII expression on monocytes from patients with Behçet's disease. Clin Immunol 2020; 219:108549. [PMID: 32739412 DOI: 10.1016/j.clim.2020.108549] [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: 04/21/2020] [Revised: 07/26/2020] [Accepted: 07/26/2020] [Indexed: 10/23/2022]
Abstract
Behçet's disease (BD) patients have abnormal FcγR polymorphisms, the implication of which remains elusive. We examined FcγRIIb expression on neutrophils, monocytes, B cells, natural killer cells, dendritic cells and T cells, and FcγRI and FcγRIII expression on monocytes in BD patients and healthy controls using flow cytometry. We further stimulated monocytes with IgG and (or) lipopolysaccharide (LPS) and measured IL-6 and TNF-α production by enzyme-linked immunosorbent assay. We found that BD monocytes expressed a lower level of FcγRIIb and a higher level of FcγRIII, which were correlated with erythrocyte sedimentation rate and C-reactive protein and were rescued after treatment. Furthermore, LPS- and IgG-stimulated BD monocytes produced higher levels of IL-6 and TNF-α than HC monocytes. In summary, we found that BD monocytes downregulated FcγRIIb expression and upregulated FcγRIII expression, which were correlated with disease activity and potentially contributed to monocyte hyperactivation in BD.
Collapse
Affiliation(s)
- Linfang Huang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China; Department of Rheumatology and Clinical Immunology, The First People's Hospital of Yueyang, Yueyang, Hunan 414000, China
| | - Xin Yu
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China
| | - Lu Li
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China
| | - Jinjing Liu
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China
| | - Xiuhua Wu
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China
| | - Yan Zeng
- Department of Rheumatology and Clinical Immunology, The First People's Hospital of Yueyang, Yueyang, Hunan 414000, China
| | - Xiangping Liao
- Department of Rheumatology and Clinical Immunology, The First People's Hospital of ChenZhou, ChenZhou, Hunan 423000, China
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Fengchun Zhang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China
| | - Xuan Zhang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China
| | - Hua Chen
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China.
| | - Wenjie Zheng
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases, Shuafuyuan, Dongcheng District, Beijing 100730, China.
| |
Collapse
|
28
|
Danzer H, Glaesner J, Baerenwaldt A, Reitinger C, Lux A, Heger L, Dudziak D, Harrer T, Gessner A, Nimmerjahn F. Human Fcγ-receptor IIb modulates pathogen-specific versus self-reactive antibody responses in lyme arthritis. eLife 2020; 9:55319. [PMID: 32613944 PMCID: PMC7438111 DOI: 10.7554/elife.55319] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022] Open
Abstract
Pathogen-specific antibody responses need to be tightly regulated to generate protective but limit self-reactive immune responses. While loss of humoral tolerance has been associated with microbial infections, the pathways involved in balancing protective versus autoreactive antibody responses in humans are incompletely understood. Studies in classical mouse model systems have provided evidence that balancing of immune responses through inhibitory receptors is an important quality control checkpoint. Genetic differences between inbred mouse models and the outbred human population and allelic receptor variants not present in mice; however, argue for caution when directly translating these findings to the human system. By studying Borrelia burgdorferi infection in humanized mice reconstituted with human hematopoietic stem cells from donors homozygous for a functional or a non-functional FcγRIIb allele, we show that the human inhibitory FcγRIIb is a critical checkpoint balancing protective and autoreactive immune responses, linking infection with induction of autoimmunity in the human immune system.
Collapse
Affiliation(s)
- Heike Danzer
- Institute of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Joachim Glaesner
- Institute of Medical Microbiology and Hygiene, University Regensburg, Regensburg, Germany
| | - Anne Baerenwaldt
- Laboratory for Cancer Immunotherapy, University Hospital Basel, Basel, Switzerland
| | - Carmen Reitinger
- Institute of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Anja Lux
- Institute of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Lukas Heger
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen, Erlangen, Germany
| | - Diana Dudziak
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen, Erlangen, Germany.,Medical Immunology Campus Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Thomas Harrer
- Medical Department 3, University Hospital Erlangen, Erlangen, Germany
| | - André Gessner
- Institute of Medical Microbiology and Hygiene, University Regensburg, Regensburg, Germany
| | - Falk Nimmerjahn
- Institute of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany.,Medical Immunology Campus Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
29
|
Gornowicz-Porowska J, Kowalczyk MJ, Seraszek-Jaros A, Bowszyc-Dmochowska M, Kaczmarek E, Żaba R, Dmochowski M. A Comparative Analysis of CD32A and CD16A Polymorphisms in Relation to Autoimmune Responses in Pemphigus Diseases and Subepithelial Autoimmune Blistering Disorders. Genes (Basel) 2020; 11:genes11040371. [PMID: 32235430 PMCID: PMC7231204 DOI: 10.3390/genes11040371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/24/2020] [Accepted: 03/28/2020] [Indexed: 12/27/2022] Open
Abstract
Autoimmune blistering dermatoses (ABDs) are characterized by autoantibodies to keratinocyte surface antigens and molecules within the dermal–epidermal junction causing disruption of skin integrity. The affinity of Fc receptors (FcRs) causing an autoimmune response in ABDs may vary based on single-nucleotide polymorphisms (SNPs) in FcRs determining the course of disease. This study aimed to explore the effects of CD16A and CD32A SNPs on the autoimmune response in several ABDs. In total, 61 ABDs patients were investigated. ELISA tests, direct immunofluorescence (DIF), TaqMan SNP Genotyping Assays, and statistical analyses were performed. The CA genotype (composed of allele C and A) of rs396991 in CD16A had a higher affinity for tissue-bound IgG1 in pemphigus and for C3 in subepithelial ABDs, showing statistical significance. The greatest relative risk (odds ratio) was reported for AA (rs396991 of CD16A) and CC (rs1801274 of CD32A) homozygotes. There were no statistically significant differences between certain genotypes and specific circulating autoantibodies (anti-DSG1, anti-DSG3 IgG in pemphigus; anti-BP180, anti-BP230 IgG) in subepithelial ABDs. Our findings indicated that rs396991 in CD16A may be of greater importance in ABDs development. Moreover, FcR polymorphisms appeared to have a greater impact on tissue-bound antibodies detected using DIF than circulating serum antibodies in ABDs.
Collapse
Affiliation(s)
- Justyna Gornowicz-Porowska
- Department of Medicinal and Cosmetic Natural Products, Poznan University of Medical Sciences, Mazowiecka 33 Street, 60-623 Poznań, Poland
- Autoimmune Blistering Dermatoses Section, Department of Dermatology, Poznan University of Medical Sciences, Przybyszewskiego 49 Street, 60-355 Poznań, Poland;
- Correspondence: (J.G.-P.); (M.J.K.); Tel.: +48-61-848-04-75 (J.G.-P.)
| | - Michał J. Kowalczyk
- Department of Dermatology and Venereology, Poznan University of Medical Sciences, Przybyszewskiego 49 Street, 60-355 Poznań, Poland;
- Correspondence: (J.G.-P.); (M.J.K.); Tel.: +48-61-848-04-75 (J.G.-P.)
| | - Agnieszka Seraszek-Jaros
- Department of Bioinformatics and Computational Biology, Poznan University of Medical Sciences, Rokietnicka 4 Street, 60-806 Poznań, Poland; (A.S.-J.); (E.K.)
| | - Monika Bowszyc-Dmochowska
- Cutaneous Histopathology and Immunopathology Section, Department of Dermatology, Poznan University of Medical Sciences, Przybyszewskiego 49 Steet, 60-355 Poznań, Poland;
| | - Elżbieta Kaczmarek
- Department of Bioinformatics and Computational Biology, Poznan University of Medical Sciences, Rokietnicka 4 Street, 60-806 Poznań, Poland; (A.S.-J.); (E.K.)
| | - Ryszard Żaba
- Department of Dermatology and Venereology, Poznan University of Medical Sciences, Przybyszewskiego 49 Street, 60-355 Poznań, Poland;
| | - Marian Dmochowski
- Autoimmune Blistering Dermatoses Section, Department of Dermatology, Poznan University of Medical Sciences, Przybyszewskiego 49 Street, 60-355 Poznań, Poland;
| |
Collapse
|
30
|
Zhang Q, Xiang L, Zaman MH, Dong W, He G, Deng GM. Predominant Role of Immunoglobulin G in the Pathogenesis of Splenomegaly in Murine Lupus. Front Immunol 2020; 10:3020. [PMID: 32082297 PMCID: PMC7005523 DOI: 10.3389/fimmu.2019.03020] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/10/2019] [Indexed: 12/27/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is characterized by high levels of autoantibodies and multiorgan tissue damage. The pathogenesis of splenomegaly in SLE remains unknown. In this study, the role of immunoglobulin G (IgG) generation and deposition in the inflammation of the spleen and associated dysfunction in SLE was investigated. In the lupus mice, we observed the development of spontaneous splenomegaly, and we found that lupus serum IgG is an important pathological factor involved in the initiation of inflammation and further germinal center (GC) and plasma cell formation. We discovered that macrophages of the splenic marginal zone are dispensable for the GC response induced by lupus IgG, but red pulp macrophages are important for GC responses. Furthermore, we found that pathogenic lupus IgG promotes inflammation and GC formation through the macrophage-mediated secretion of TNF-α. Syk inhibitor treatment suppressed the changes in the histopathology of the spleen induced by lupus IgG. This study will contribute to the understanding of the pathogenesis of splenomegaly in lupus and promote the development of an effective therapeutic strategy for SLE.
Collapse
Affiliation(s)
- Qian Zhang
- Key Laboratory of Antibody Techniques, National Health Commission, Nanjing Medical University, Nanjing, China
| | - Liping Xiang
- Key Laboratory of Antibody Techniques, National Health Commission, Nanjing Medical University, Nanjing, China.,Department of Clinical Laboratory, Nanjing Jiangning Hospital, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Muhammad Haidar Zaman
- Key Laboratory of Antibody Techniques, National Health Commission, Nanjing Medical University, Nanjing, China
| | - Wenhui Dong
- Key Laboratory of Antibody Techniques, National Health Commission, Nanjing Medical University, Nanjing, China
| | - Guodan He
- Key Laboratory of Antibody Techniques, National Health Commission, Nanjing Medical University, Nanjing, China
| | - Guo-Min Deng
- Department of Rheumatology, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Lee WS, Amengual O. B cells targeting therapy in the management of systemic lupus erythematosus. Immunol Med 2019; 43:16-35. [PMID: 32107989 DOI: 10.1080/25785826.2019.1698929] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease which affects the majority of organs and systems. Traditional therapies do not lead to complete remission of disease but only relieve symptoms and inflammation. B cells are the most important effector cell types in the pathogenesis of SLE. Therefore, therapies targeting B cells and their related cytokines are a very important milestone for SLE treatment. Several biologics that modulate B cells, either depleting B cells or blocking B cell functions, have been developed and evaluated in clinical trials. Belimumab, a fully humanized monoclonal antibody that specifically binds B cells activating factor (BAFF), was the first of these agents approved for SLE treatment. In this review, we explore the currently available evidence in B cell targeted therapies in SLE including agents that target B cell surface antigens (CD19, CD20, CD22), B cell survival factors (BAFF and a proliferation-inducing ligand, APRIL), cytokines (interleukin-1 and type 1 interferons) and co-stimulatory molecules (CD40 ligand). We highlighted the mechanisms of action and the individual characteristics of these biologics, and present an update on the clinical trials that have evaluated their efficacy and safety. Finally, we describe some of the emerging and promising therapies for SLE treatment.
Collapse
Affiliation(s)
- Wen Shi Lee
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Olga Amengual
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
32
|
Murphy KA, Bhamidipati K, Rubin SJS, Kipp L, Robinson WH, Lanz TV. Immunomodulatory receptors are differentially expressed in B and T cell subsets relevant to autoimmune disease. Clin Immunol 2019; 209:108276. [PMID: 31669582 DOI: 10.1016/j.clim.2019.108276] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 12/15/2022]
Abstract
Inhibitory cell-surface receptors on lymphocytes, often called immune checkpoints, are powerful targets for cancer therapy. Despite their direct involvement in autoimmune pathology, they are currently not exploited therapeutically for autoimmune diseases. Understanding the expression pattern of these receptors in health and disease is essential for targeted drug design. Here, we designed three 23-colour flow cytometry panels for peripheral-blood T cells, including 15 lineage-defining markers and 21 immunomodulatory cell-surface receptors, and a 22-marker panel for B cells. Blood samples from healthy individuals, multiple sclerosis (MS), and lupus (SLE) patients were included in the study. Several receptors show differential expression on regulatory T cells (Treg) compared to T helper (Th) 1 and Th17 cells, and functional relevance of this difference could be shown for BTLA and CD5. Unbiased multiparametric analysis revealed a subset of activated CD8+ T cells and a subset of unswitched memory B cells that are diminished in MS and SLE, respectively.
Collapse
Affiliation(s)
- Katherine A Murphy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Kartik Bhamidipati
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Samuel J S Rubin
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Lucas Kipp
- Division of Neuroimmunology, Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - William H Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States.
| | - Tobias V Lanz
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States; Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
33
|
Verbeek JS, Hirose S, Nishimura H. The Complex Association of FcγRIIb With Autoimmune Susceptibility. Front Immunol 2019; 10:2061. [PMID: 31681256 PMCID: PMC6803437 DOI: 10.3389/fimmu.2019.02061] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 08/15/2019] [Indexed: 12/20/2022] Open
Abstract
FcγRIIb is the only inhibitory Fc receptor and controls many aspects of immune and inflammatory responses. The observation 19 years ago that Fc γ RIIb -/- mice generated by gene targeting in 129 derived ES cells developed severe lupus like disease when backcrossed more than 7 generations into C57BL/6 background initiated extensive research on the functional understanding of this strong autoimmune phenotype. The genomic region in the distal part of Chr1 both in human and mice in which the Fc γ R gene cluster is located shows a high level of complexity in relation to the susceptibility to SLE. Specific haplotypes of closely linked genes including the Fc γ RIIb and Slamf genes are associated with increased susceptibility to SLE both in mice and human. Using forward and reverse genetic approaches including in human GWAS and in mice congenic strains, KO mice (germline and cell type specific, on different genetic background), knockin mice, overexpressing transgenic mice combined with immunological models such as adoptive transfer of B cells from Ig transgenic mice the involved genes and the causal mutations and their associated functional alterations were analyzed. In this review the results of this 19 years extensive research are discussed with a focus on (genetically modified) mouse models.
Collapse
Affiliation(s)
- J Sjef Verbeek
- Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| | - Sachiko Hirose
- Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| | - Hiroyuki Nishimura
- Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| |
Collapse
|
34
|
Lu KL, Wu MY, Wang CH, Wang CW, Hung SI, Chung WH, Chen CB. The Role of Immune Checkpoint Receptors in Regulating Immune Reactivity in Lupus. Cells 2019; 8:E1213. [PMID: 31597242 PMCID: PMC6829486 DOI: 10.3390/cells8101213] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 09/29/2019] [Accepted: 10/03/2019] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint receptors with co-stimulatory and co-inhibitory signals are important modulators for the immune system. However, unrestricted co-stimulation and/or inadequate co-inhibition may cause breakdown of self-tolerance, leading to autoimmunity. Systemic lupus erythematosus (SLE) is a complex multi-organ disease with skewed and dysregulated immune responses interacting with genetics and the environment. The close connections between co-signaling pathways and SLE have gradually been established in past research. Also, the recent success of immune checkpoint blockade in cancer therapy illustrates the importance of the co-inhibitory receptors in cancer immunotherapy. Moreover, immune checkpoint blockade could result in substantial immune-related adverse events that mimic autoimmune diseases, including lupus. Together, immune checkpoint regulators represent viable immunotherapeutic targets for the treatment of both autoimmunity and cancer. Therefore, it appears reasonable to treat SLE by restoring the out-of-order co-signaling axis or by manipulating collateral pathways to control the pathogenic immune responses. Here, we review the current state of knowledge regarding the relationships between SLE and the co-signaling pathways of T cells, B cells, dendritic cells, and neutrophils, and highlight their potential clinical implications. Current clinical trials targeting the specific co-signaling axes involved in SLE help to advance such knowledge, but further in-depth exploration is still warranted.
Collapse
Affiliation(s)
- Kun-Lin Lu
- Chang Gung Memorial Hospital, Linkou 333, Taiwan; (K.-L.L.); (M.-Y.W.); , (C.-W.W.); (S.-I.H.)
- College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan
| | - Ming-Ying Wu
- Chang Gung Memorial Hospital, Linkou 333, Taiwan; (K.-L.L.); (M.-Y.W.); , (C.-W.W.); (S.-I.H.)
- College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei 105, Taiwan
| | - Chi-Hui Wang
- Chang Gung Memorial Hospital, Linkou 333, Taiwan; (K.-L.L.); (M.-Y.W.); , (C.-W.W.); (S.-I.H.)
- College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei 105, Taiwan
| | - Chuang-Wei Wang
- Chang Gung Memorial Hospital, Linkou 333, Taiwan; (K.-L.L.); (M.-Y.W.); , (C.-W.W.); (S.-I.H.)
- College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei 105, Taiwan
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Linkou 333, Taiwan
| | - Shuen-Iu Hung
- Chang Gung Memorial Hospital, Linkou 333, Taiwan; (K.-L.L.); (M.-Y.W.); , (C.-W.W.); (S.-I.H.)
- College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei 105, Taiwan
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Linkou 333, Taiwan
| | - Wen-Hung Chung
- Chang Gung Memorial Hospital, Linkou 333, Taiwan; (K.-L.L.); (M.-Y.W.); , (C.-W.W.); (S.-I.H.)
- College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei 105, Taiwan
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Linkou 333, Taiwan
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung 204, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou 333, Taiwan
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen 361000, China
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan
| | - Chun-Bing Chen
- Chang Gung Memorial Hospital, Linkou 333, Taiwan; (K.-L.L.); (M.-Y.W.); , (C.-W.W.); (S.-I.H.)
- College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei 105, Taiwan
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Linkou 333, Taiwan
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung 204, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou 333, Taiwan
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen 361000, China
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan
| |
Collapse
|
35
|
Nagelkerke SQ, Schmidt DE, de Haas M, Kuijpers TW. Genetic Variation in Low-To-Medium-Affinity Fcγ Receptors: Functional Consequences, Disease Associations, and Opportunities for Personalized Medicine. Front Immunol 2019; 10:2237. [PMID: 31632391 PMCID: PMC6786274 DOI: 10.3389/fimmu.2019.02237] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/04/2019] [Indexed: 12/23/2022] Open
Abstract
Fc-gamma receptors (FcγR) are the cellular receptors for Immunoglobulin G (IgG). Upon binding of complexed IgG, FcγRs can trigger various cellular immune effector functions, thereby linking the adaptive and innate immune systems. In humans, six classic FcγRs are known: one high-affinity receptor (FcγRI) and five low-to-medium-affinity FcγRs (FcγRIIA, -B and -C, FcγRIIIA and -B). In this review we describe the five genes encoding the low-to-medium -affinity FcγRs (FCGR2A, FCGR2B, FCGR2C, FCGR3A, and FCGR3B), including well-characterized functionally relevant single nucleotide polymorphisms (SNPs), haplotypes as well as copy number variants (CNVs), which occur in distinct copy number regions across the locus. The evolution of the locus is also discussed. Importantly, we recommend a consistent nomenclature of genetic variants in the FCGR2/3 locus. Next, we focus on the relevance of genetic variation in the FCGR2/3 locus in auto-immune and auto-inflammatory diseases, highlighting pathophysiological insights that are informed by genetic association studies. Finally, we illustrate how specific FcγR variants relate to variation in treatment responses and prognosis amongst autoimmune diseases, cancer and transplant immunology, suggesting novel opportunities for personalized medicine.
Collapse
Affiliation(s)
- Sietse Q Nagelkerke
- Sanquin Research and Landsteiner Laboratory, Department of Blood Cell Research, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - David E Schmidt
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Masja de Haas
- Sanquin Diagnostic Services, Department of Immunohematology Diagnostics, Amsterdam, Netherlands.,Sanquin Research, Center for Clinical Transfusion Research, Leiden, Netherlands.,Jon J. van Rood Center for Clinical Transfusion Science, Leiden University Medical Center, Leiden, Netherlands.,Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Taco W Kuijpers
- Sanquin Research and Landsteiner Laboratory, Department of Blood Cell Research, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
36
|
Weißenberg SY, Szelinski F, Schrezenmeier E, Stefanski AL, Wiedemann A, Rincon-Arevalo H, Welle A, Jungmann A, Nordström K, Walter J, Imgenberg-Kreuz J, Nordmark G, Rönnblom L, Bachali P, Catalina MD, Grammer AC, Lipsky PE, Lino AC, Dörner T. Identification and Characterization of Post-activated B Cells in Systemic Autoimmune Diseases. Front Immunol 2019; 10:2136. [PMID: 31616406 PMCID: PMC6768969 DOI: 10.3389/fimmu.2019.02136] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/27/2019] [Indexed: 12/16/2022] Open
Abstract
Autoimmune diseases (AID) such as systemic lupus erythematosus (SLE), primary Sjögren's syndrome (pSS), and rheumatoid arthritis (RA) are chronic inflammatory diseases in which abnormalities of B cell function play a central role. Although it is widely accepted that autoimmune B cells are hyperactive in vivo, a full understanding of their functional status in AID has not been delineated. Here, we present a detailed analysis of the functional capabilities of AID B cells and dissect the mechanisms underlying altered B cell function. Upon BCR activation, decreased spleen tyrosine kinase (Syk) and Bruton's tyrosine kinase (Btk) phosphorylation was noted in AID memory B cells combined with constitutive co-localization of CD22 and protein tyrosine phosphatase (PTP) non-receptor type 6 (SHP-1) along with hyporesponsiveness to TLR9 signaling, a Syk-dependent response. Similar BCR hyporesponsiveness was also noted specifically in SLE CD27− B cells together with increased PTP activities and increased transcripts for PTPN2, PTPN11, PTPN22, PTPRC, and PTPRO in SLE B cells. Additional studies revealed that repetitive BCR stimulation of normal B cells can induce BCR hyporesponsiveness and that tissue-resident memory B cells from AID patients also exhibited decreased responsiveness immediately ex vivo, suggesting that the hyporesponsive status can be acquired by repeated exposure to autoantigen(s) in vivo. Functional studies to overcome B cell hyporesponsiveness revealed that CD40 co-stimulation increased BCR signaling, induced proliferation, and downregulated PTP expression (PTPN2, PTPN22, and receptor-type PTPs). The data support the conclusion that hyporesponsiveness of AID and especially SLE B cells results from chronic in vivo stimulation through the BCR without T cell help mediated by CD40–CD154 interaction and is manifested by decreased phosphorylation of BCR-related proximal signaling molecules and increased PTPs. The hyporesponsiveness of AID B cells is similar to a form of functional anergy.
Collapse
Affiliation(s)
- Sarah Y Weißenberg
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany.,German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Franziska Szelinski
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany.,German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Eva Schrezenmeier
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany
| | - Ana-Luisa Stefanski
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany
| | - Annika Wiedemann
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany
| | - Hector Rincon-Arevalo
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany.,German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany.,Grupo de Inmunología Celular e Inmunogenética, Facultad de Medicina, Instituto de Investigaciones Médicas, Universidad de Antioquia UdeA, Medellín, Colombia
| | - Anna Welle
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken, Germany
| | - Annemarie Jungmann
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken, Germany
| | - Karl Nordström
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken, Germany
| | - Jörn Walter
- Department of Genetics and Epigenetics, Saarland University, Saarbrücken, Germany
| | - Juliana Imgenberg-Kreuz
- Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Gunnel Nordmark
- Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lars Rönnblom
- Department of Medical Sciences, Rheumatology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | | | - Amrie C Grammer
- RILITE Research Institute, Charlottesville, VA, United States
| | - Peter E Lipsky
- RILITE Research Institute, Charlottesville, VA, United States
| | - Andreia C Lino
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany.,German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Thomas Dörner
- Department of Rheumatology and Clinical Immunology, Charité University Medicine Berlin, Berlin, Germany.,German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| |
Collapse
|
37
|
Wang Y, Jönsson F. Expression, Role, and Regulation of Neutrophil Fcγ Receptors. Front Immunol 2019; 10:1958. [PMID: 31507592 PMCID: PMC6718464 DOI: 10.3389/fimmu.2019.01958] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/02/2019] [Indexed: 12/31/2022] Open
Abstract
Neutrophils are best known for their critical role in host defense, for which they utilize multiple innate immune mechanisms, including microbe-associated pattern recognition, phagocytosis, production of reactive oxygen species, and the release of potent proteases, mediators, antimicrobials, and neutrophil extracellular traps. Beyond their well-established contribution to innate immunity, neutrophils were more recently reported to interact with various other cell types, including cells from the adaptive immune system, thereby enabling neutrophils to tune the overall immune response of the host. Neutrophils express different receptors for IgG antibodies (Fcγ receptors), which facilitate the engulfment of IgG-opsonized microbes and trigger cell activation upon cross-linking of several receptors. Indeed, FcγRs (via IgG antibodies) confer neutrophils with a key feature of the adaptive immunity: an antigen-specific cell response. This review summarizes the expression and function of FcγRs on human neutrophils in health and disease and how they are affected by polymorphisms in the FCGR loci. Additionally, we will discuss the role of neutrophils in providing help to marginal zone B cells for the production of antibodies, which in turn may trigger neutrophil effector functions when engaging FcγRs.
Collapse
Affiliation(s)
- Yu Wang
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR 1222 INSERM, Paris, France
- Université Diderot Paris VII, PSL University, Paris, France
| | - Friederike Jönsson
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR 1222 INSERM, Paris, France
| |
Collapse
|
38
|
Lu X, Peng S, Wang X, Shan Z, Teng W. Decreased expression of FcγRII in active Graves' disease patients. J Clin Lab Anal 2019; 33:e22904. [PMID: 31033004 PMCID: PMC6642309 DOI: 10.1002/jcla.22904] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/27/2019] [Accepted: 03/28/2019] [Indexed: 12/31/2022] Open
Abstract
Background Graves' disease (GD) is a common autoimmune disease characterized by genetic and environmental factors. Fcγ receptors (FcγRs) are involved in several autoimmune disorders through recognizing immunoglobulin (Ig) G antibodies and mediating immune response. The study on the expression of FcγRs in GD patients is scarce. The purpose of this study was to evaluate the expression of three different types of FcγRs in patients with active and remissive GD. Methods Blood samples of patients and healthy subjects were collected to analyze the percentage of FcγRI (CD64), FcγRII (CD32), and FcγRIII (CD16) on peripheral blood mononuclear cells (PBMCs) and monocytes by flow cytometry and Western blotting. CD32 isotypes were also examined in cases and controls by real‐time PCR. Results The cell percentages expressed CD32 and protein expressions of CD32 on PBMCs, and monocytes from patients with active GD were significantly reduced compared to controls and patients with remissive GD. In particular, the expression of CD32B on PBMC was also decreased in active GD patients. However, the cell percentages expressed CD16 and CD64 from PBMCs and monocytes were comparable between three groups. Besides, the percentages of CD14+CD32+ cells were negatively correlated with TRAb titers in active GD patients (r = −0.5825, P < 0.001). Conclusion These results suggested that CD32 may act as a novel marker for active GDs. The expression of monocytic CD32, in particular CD32B, in GD patients might play a crucial role in maintaining FcγRs function and be a therapeutic target in GD patients.
Collapse
Affiliation(s)
- Xixuan Lu
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shiqiao Peng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xinyi Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhongyan Shan
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Weiping Teng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, Liaoning Provincial Key Laboratory of Endocrine Diseases, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
39
|
Ben Mkaddem S, Benhamou M, Monteiro RC. Understanding Fc Receptor Involvement in Inflammatory Diseases: From Mechanisms to New Therapeutic Tools. Front Immunol 2019; 10:811. [PMID: 31057544 PMCID: PMC6481281 DOI: 10.3389/fimmu.2019.00811] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 03/27/2019] [Indexed: 12/21/2022] Open
Abstract
Fc receptors (FcRs) belong to the ITAM-associated receptor family. FcRs control the humoral and innate immunity which are essential for appropriate responses to infections and prevention of chronic inflammation or auto-immune diseases. Following their crosslinking by immune complexes, FcRs play various roles such as modulation of the immune response by released cytokines or of phagocytosis. Here, we review FcR involvement in pathologies leading notably to altered intracellular signaling with functionally relevant consequences to the host, and targeting of Fc receptors as therapeutic approaches. Special emphasis will be given to some FcRs, such as the FcαRI, the FcγRIIA and the FcγRIIIA, which behave like the ancient god Janus depending on the ITAM motif to inhibit or activate immune responses depending on their targeting by monomeric/dimeric immunoglobulins or by immune complexes. This ITAM duality has been recently defined as inhibitory or activating ITAM (ITAMi or ITAMa) which are controlled by Src family kinases. Involvement of various ITAM-bearing FcRs observed during infectious or autoimmune diseases is associated with allelic variants, changes in ligand binding ability responsible for host defense perturbation. During auto-immune diseases such as rheumatoid arthritis, lupus or immune thrombocytopenia, the autoantibodies and immune complexes lead to inflammation through FcR aggregation. We will discuss the role of FcRs in autoimmune diseases, and focus on novel approaches to target FcRs for resolution of antibody-mediated autoimmunity. We will finally also discuss the down-regulation of FcR functionality as a therapeutic approach for autoimmune diseases.
Collapse
Affiliation(s)
- Sanae Ben Mkaddem
- INSERM U1149, Centre de Recherche sur l'Inflammation, Paris, France.,CNRS ERL8252, Paris, France.,Faculté de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Site Xavier Bichat, Paris, France.,Inflamex Laboratory of Excellence, Paris, France
| | - Marc Benhamou
- INSERM U1149, Centre de Recherche sur l'Inflammation, Paris, France.,CNRS ERL8252, Paris, France.,Faculté de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Site Xavier Bichat, Paris, France.,Inflamex Laboratory of Excellence, Paris, France
| | - Renato C Monteiro
- INSERM U1149, Centre de Recherche sur l'Inflammation, Paris, France.,CNRS ERL8252, Paris, France.,Faculté de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Site Xavier Bichat, Paris, France.,Inflamex Laboratory of Excellence, Paris, France.,Service d'Immunologie, DHU Fire, Hôpital Bichat-Claude Bernard, Assistance Publique de Paris, Paris, France
| |
Collapse
|
40
|
Anania JC, Chenoweth AM, Wines BD, Hogarth PM. The Human FcγRII (CD32) Family of Leukocyte FcR in Health and Disease. Front Immunol 2019; 10:464. [PMID: 30941127 PMCID: PMC6433993 DOI: 10.3389/fimmu.2019.00464] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/21/2019] [Indexed: 12/15/2022] Open
Abstract
FcγRs have been the focus of extensive research due to their key role linking innate and humoral immunity and their implication in both inflammatory and infectious disease. Within the human FcγR family FcγRII (activatory FcγRIIa and FcγRIIc, and inhibitory FcγRIIb) are unique in their ability to signal independent of the common γ chain. Through improved understanding of the structure of these receptors and how this affects their function we may be able to better understand how to target FcγR specific immune activation or inhibition, which will facilitate in the development of therapeutic monoclonal antibodies in patients where FcγRII activity may be desirable for efficacy. This review is focused on roles of the human FcγRII family members and their link to immunoregulation in healthy individuals and infection, autoimmunity and cancer.
Collapse
Affiliation(s)
- Jessica C Anania
- Centre for Biomedical Research, Burnet Institute, Melbourne, VIC, Australia.,Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Alicia M Chenoweth
- Centre for Biomedical Research, Burnet Institute, Melbourne, VIC, Australia.,Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Bruce D Wines
- Centre for Biomedical Research, Burnet Institute, Melbourne, VIC, Australia.,Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - P Mark Hogarth
- Centre for Biomedical Research, Burnet Institute, Melbourne, VIC, Australia.,Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
41
|
Saiworn W, Thim-Uam A, Visitchanakun P, Atjanasuppat K, Chantaraaumporn J, Mokdara J, Chungchatupornchai S, Pisitkun P, Leelahavanichkul A, Poolthong S, Baron R, Lotinun S. Cortical Bone Loss in a Spontaneous Murine Model of Systemic Lupus Erythematosus. Calcif Tissue Int 2018; 103:686-697. [PMID: 30116830 DOI: 10.1007/s00223-018-0464-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 08/08/2018] [Indexed: 01/10/2023]
Abstract
Patients with systemic lupus erythematosus (SLE), a chronic inflammatory disease characterized by loss of T- and B-cell tolerance to autoantigens, are at increased risk for osteoporosis and fractures. Mice deficient in Fc gamma receptor IIb (FcγRIIB) exhibit spontaneous SLE and its restoration rescues the disease. To determine whether deleting FcγRIIB affects cortical bone mass and mechanical properties, we analyzed cortical bone phenotype of FcγRIIB knockouts at different ages. FACS analysis revealed that 6-month-old FcγRIIB-/- mice had increased B220lowCD138+ cells, markers of plasma cells, indicating active SLE disease. In contrast, 3-month-old FcγRIIB-/- mice did not develop the active SLE disease. µCT analysis indicated that FcγRIIB deletion did not affect cortical bone in 3-month-old mutants. However, 6- and 10-month-old FcγRIIB-/- males and females had osteopenic cortical bone and the severity of bone loss increased with disease duration. FcγRIIB deletion decreased cross-sectional area, cortical area, and marrow area in 6-month-old males. Cortical area and cortical thickness were decreased in 10-month-old FcγRIIB-/- males. Lack of FcγRIIB decreased cortical thickness without affecting cortical area in females. However, deletion of a single FcγRIIB allele was insufficient to induce cortical bone loss. The bending strength was decreased in 6- and 10-month-old FcγRIIB-deficient males compared to WT controls. A microindentation analysis demonstrated significantly decreased hardness in both 10-month-old FcγRIIB-/- males and females. Our data indicate that FcγRIIB contributes to the regulation of cortical bone homeostasis subsequent to SLE development and that deletion of FcγRIIB in mice leads to SLE-like disease associated with cortical bone loss and decreased bending strength and hardness.
Collapse
Affiliation(s)
- Worasit Saiworn
- Department of Physiology and Skeletal Disorders Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Arthid Thim-Uam
- Inter-Department Program of Biomedical Sciences, Faculty of Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Peerapat Visitchanakun
- Department of Physiology and Skeletal Disorders Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Korakot Atjanasuppat
- Department of Physiology and Skeletal Disorders Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Jiratha Chantaraaumporn
- Department of Physiology and Skeletal Disorders Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Jutarat Mokdara
- Department of Physiology and Skeletal Disorders Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Sirintra Chungchatupornchai
- Department of Physiology and Skeletal Disorders Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Prapaporn Pisitkun
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Division of Immunology, Department of Microbiology, Faculty of Medicine, and Skeletal Disorders Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Suchit Poolthong
- Department of Prosthodontics and Skeletal Disorders Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Roland Baron
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
- Harvard Medical School, Department of Medicine, Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Sutada Lotinun
- Department of Physiology and Skeletal Disorders Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
42
|
Cis interaction between sialylated FcγRIIA and the αI-domain of Mac-1 limits antibody-mediated neutrophil recruitment. Nat Commun 2018; 9:5058. [PMID: 30498196 PMCID: PMC6265255 DOI: 10.1038/s41467-018-07506-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 11/07/2018] [Indexed: 12/14/2022] Open
Abstract
Vascular-deposited IgG immune complexes promote neutrophil recruitment, but how this process is regulated is still unclear. Here we show that the CD18 integrin Mac-1, in its bent state, interacts with the IgG receptor FcγRIIA in cis to reduce the affinity of FcγRIIA for IgG and inhibit FcγRIIA-mediated neutrophil recruitment under flow. The Mac-1 rs1143679 lupus-risk variant reverses Mac-1 inhibition of FcγRIIA, as does a Mac-1 ligand and a mutation in Mac-1’s ligand binding αI-domain. Sialylated complex glycans on FcγRIIA interact with the αI-domain via divalent cations, and this interaction is required for FcγRIIA inhibition by Mac-1. Human neutrophils deficient in CD18 integrins exhibit augmented FcγRIIA-dependent recruitment to IgG-coated endothelium. In mice, CD18 integrins on neutrophils dampen IgG-mediated neutrophil accumulation in the kidney. In summary, cis interaction between sialylated FcγRIIA and the αI-domain of Mac-1 alters the threshold for IgG-mediated neutrophil recruitment. A disruption of this interaction may increase neutrophil influx in autoimmune diseases. Deposited immune complexes (IC) promote neutrophil recruitment, but the fine tuning of this process is still unclear. Here the authors show that the cis interaction of the IC receptor, FcγRIIA and CD18 integrin, Mac-1, on the neutrophil surface modulates neutrophil adhesion, with FcγRIIA sialylation specifically implicated in this interaction.
Collapse
|
43
|
Williams V, Rawat A, Vignesh P, Shandilya JK, Gupta A, Singh S. Fc-gamma receptor expression profile in a North-Indian cohort of pediatric-onset systemic lupus erythematosus: An observational study. Int J Rheum Dis 2018; 22:449-457. [PMID: 30399647 DOI: 10.1111/1756-185x.13403] [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: 12/03/2017] [Revised: 07/11/2018] [Accepted: 09/14/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND Polymorphisms in the Fcγ-receptor (FcγR) have been associated with increased susceptibility to systemic lupus erythematosus (SLE). There is a paucity of data on FcγR expression pattern in pediatric subjects with SLE. The aim of the study was to assess the expression of various FcγRs by flow cytometry in children with pediatric-onset SLE (pSLE). METHODS Thirty-one children aged 0-15 years fulfilling 2012 Systemic Lupus International Collaborating Clinics Classification Criteria for SLE were enrolled. Disease-active (n = 14) and the inactive group were delineated using the SLE Disease Activity Index (SLEDAI). Thirteen age- and sex-matched controls were also enrolled. Blood samples of cases and controls were assessed for CD64, CD32B and CD16 expression on B lymphocytes, neutrophils and monocytes by flow cytometry using standard techniques. Median fluorescence intensity (MFI) and percentage expression were calculated using the FACS DIVA software and Kaluza software. RESULTS Median fluorescence intensity and percentage expression of CD64 on monocytes (MFI: 1.71 vs 1.51, P = 0.86) and neutrophils (MFI: 0.42 vs 0.64, P = 0.3) were comparable between patients and controls. MFIs of CD16 expression on neutrophils (3.47 vs 11.4, P = 0.05) and monocytes (1.28 vs 3.45, P = 0.07) were lower in patients compared to controls. CD32B expression on lymphocytes (MFI: 0.56 vs 1.37; % expression:18.3% vs 12.32%) was also comparable between cases and controls. Expression of CD64, CD16, and CD32B were also comparable between patients with active and inactive disease. CONCLUSION No significant differences were observed in FcγR expression between patients and controls. However, the overall trends of FcγR expression and decreased CD16 on monocytes and neutrophils are in consonance with data from larger cohorts of adult SLE patients.
Collapse
Affiliation(s)
- Vijai Williams
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Amit Rawat
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Pandiarajan Vignesh
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Jitendra Kumar Shandilya
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Anju Gupta
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Surjit Singh
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
44
|
Jhou JP, Yu IS, Hwai H, Chen CS, Chen PL, Tzeng SJ. The Lupus-Associated Fcγ Receptor IIb-I232T Polymorphism Results in Impairment in the Negative Selection of Low-Affinity Germinal Center B Cells Via c-Abl in Mice. Arthritis Rheumatol 2018; 70:1866-1878. [PMID: 29774664 PMCID: PMC6221021 DOI: 10.1002/art.40555] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 05/08/2018] [Indexed: 11/17/2022]
Abstract
Objective Fcγ receptor IIb (FcγRIIb) is an essential negative regulator of B cells that blocks B cell receptor (BCR) signaling and triggers c‐Abl–dependent apoptosis of B cells. FcγRIIb‐deficient mice display splenomegaly with expansion of B cells, leading to lupus. FcγRIIb‐I232T is a hypofunctional polymorphism associated with lupus susceptibility in humans, an autoimmune disease linked to diminished deletion of autoreactive B cells. In the context of the FcγRIIb‐I232T polymorphism, we investigated the role of FcγRIIb in the deletion of low‐affinity germinal center (GC) B cells, an important mechanism for preventing autoimmunity. Methods We generated FcγRIIb232T/T mice to mimic human FcγRIIb‐I232T carriers and immunized mice with chicken gamma globulin (CGG)–conjugated NP, a T cell–dependent antigen, to examine the response of GC B cells. Results Compared to wild‐type (WT) mice, FcγRIIb232T/T mice showed increased numbers of low‐affinity NP‐specific IgG and NP‐specific B cells and plasma cells; additionally, the expression of a somatic mutation (W33L) in their VH186.2 genes encoding high‐affinity BCR was reduced. Notably, FcγRIIb232T/T mice had a higher number of GC light zone B cells and showed less apoptosis than WT mice, despite having equivalent follicular helper T cell numbers and function. Moreover, phosphorylation of c‐Abl was reduced in FcγRIIb232T/T mice, and treatment of WT mice with the c‐Abl inhibitor nilotinib during the peak of GC response resulted in reduced affinity maturation reminiscent of FcγRIIb232T/T mice. Conclusion Our findings provide evidence of a critical role of FcγRIIb/c‐Abl in the negative selection of GC B cells in FcγRIIb232T/T mice. Importantly, our findings indicate potential benefits of up‐regulating FcγRIIb expression in B cells for treatment of systemic lupus erythematosus.
Collapse
Affiliation(s)
| | - I-Shing Yu
- National Taiwan University, Taipei, Taiwan
| | - Haw Hwai
- National Taiwan University, Taipei, Taiwan
| | | | | | | |
Collapse
|
45
|
Wines BD, Billings H, Mclean MR, Kent SJ, Hogarth PM. Antibody Functional Assays as Measures of Fc Receptor-Mediated Immunity to HIV - New Technologies and their Impact on the HIV Vaccine Field. Curr HIV Res 2018; 15:202-215. [PMID: 28322167 PMCID: PMC5543561 DOI: 10.2174/1570162x15666170320112247] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 02/20/2017] [Accepted: 03/09/2017] [Indexed: 12/23/2022]
Abstract
Background: There is now intense interest in the role of HIV-specific antibodies and the engagement of FcγR functions in the control and prevention of HIV infection. The analyses of the RV144 vaccine trial, natural progression cohorts, and macaque models all point to a role for Fc-dependent effector functions, such as cytotoxicity (ADCC) or phagocytosis (ADCP), in the control of HIV. However, reliable assays that can be reproducibly used across different laboratories to measure Fc-dependent functions, such as antibody dependent cellular cytotoxicity (ADCC) are limited. Method: This brief review highlights the importance of Fc properties for immunity to HIV, particular-ly via FcγR diversity and function. We discuss assays used to study FcR mediated functions of HIV-specific Ab, including our recently developed novel cell-free ELISA using homo-dimeric FcγR ecto-domains to detect functionally relevant viral antigen-specific antibodies. Results: The binding of these dimeric FcγR ectodomains, to closely spaced pairs of IgG Fc, mimics the engagement and cross-linking of Fc receptors by IgG opsonized virions or infected cells as the es-sential prerequisite to the induction of Ab-dependent effector functions. The dimeric FcγR ELISA reli-ably correlates with ADCC in patient responses to influenza. The assay is amenable to high throughput and could be standardized across laboratories. Conclusion: We propose the assay has broader implications for the evaluation of the quality of anti-body responses in viral infections and for the rapid evaluation of responses in vaccine development campaigns for HIV and other viral infections.
Collapse
Affiliation(s)
- Bruce D Wines
- Centre for Biomedical Research, Burnet Institute, Melbourne, Vic 3004, Australia.,Department of Immunology, Monash University Central Clinical School, Melbourne, Victoria 3004, Australia.,Department of Pathology, The University of Melbourne, Victoria, 3010, Australia
| | - Hugh Billings
- Centre for Biomedical Research, Burnet Institute, Melbourne, Vic 3004, Australia
| | - Milla R Mclean
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Parkville, Victoria, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Parkville, Victoria, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Melbourne, Parkville, Victoria, Australia.,Melbourne Sexual Health Centre, Infectious Diseases Department, Alfred Health, Central Clinical School, Monash University, Victoria, Australia
| | - P Mark Hogarth
- Centre for Biomedical Research, Burnet Institute, Melbourne, Vic 3004, Australia.,Department of Immunology, Monash University Central Clinical School, Melbourne, Victoria 3004, Australia.,Department of Pathology, The University of Melbourne, Victoria, 3010, Australia
| |
Collapse
|
46
|
Jimenez RV, Wright TT, Jones NR, Wu J, Gibson AW, Szalai AJ. C-Reactive Protein Impairs Dendritic Cell Development, Maturation, and Function: Implications for Peripheral Tolerance. Front Immunol 2018; 9:372. [PMID: 29556231 PMCID: PMC5845098 DOI: 10.3389/fimmu.2018.00372] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 02/09/2018] [Indexed: 12/23/2022] Open
Abstract
C-reactive protein (CRP) is the prototypical acute phase reactant, increasing in blood concentration rapidly and several-fold in response to inflammation. Recent evidence indicates that CRP has an important physiological role even at low, baseline levels, or in the absence of overt inflammation. For example, we have shown that human CRP inhibits the progression of experimental autoimmune encephalomyelitis (EAE) in CRP transgenic mice by shifting CD4+ T cells away from the TH1 and toward the TH2 subset. Notably, this action required the inhibitory Fcγ receptor IIB (FcγRIIB), but did not require high levels of human CRP. Herein, we sought to determine if CRP's influence in EAE might be explained by CRP acting on dendritic cells (DC; antigen presenting cells known to express FcγRIIB). We found that CRP (50 µg/ml) reduced the yield of CD11c+ bone marrow-derived DCs (BMDCs) and CRP (≥5 μg/ml) prevented their full expression of major histocompatibility complex class II and the co-stimulatory molecules CD86 and CD40. CRP also decreased the ability of BMDCs to stimulate antigen-driven proliferation of T cells in vitro. Importantly, if the BMDCs were genetically deficient in mouse FcγRIIB then (i) the ability of CRP to alter BMDC surface phenotype and impair T cell proliferation was ablated and (ii) CD11c-driven expression of a human FCGR2B transgene rescued the CRP effect. Lastly, the protective influence of CRP in EAE was fully restored in mice with CD11c-driven human FcγRIIB expression. These findings add to the growing evidence that CRP has important biological effects even in the absence of an acute phase response, i.e., CRP acts as a tonic suppressor of the adaptive immune system. The ability of CRP to suppress development, maturation, and function of DCs implicates CRP in the maintenance of peripheral T cell tolerance.
Collapse
Affiliation(s)
- Rachel V. Jimenez
- Department of Medicine, Division of Clinical Immunology & Rheumatology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Tyler T. Wright
- Division of Drug Development, Southern Research, Birmingham, AL, United States
| | - Nicholas R. Jones
- Department of Medicine, Division of Clinical Immunology & Rheumatology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jianming Wu
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Minneapolis, MN, United States
| | - Andrew W. Gibson
- Department of Medicine, Division of Clinical Immunology & Rheumatology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Alexander J. Szalai
- Department of Medicine, Division of Clinical Immunology & Rheumatology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
47
|
Fc Gamma Receptor IIB Deficient Mice: A Lupus Model with Increased Endotoxin Tolerance-Related Sepsis Susceptibility. Shock 2018; 47:743-752. [PMID: 27849678 DOI: 10.1097/shk.0000000000000796] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hyper-elevated immune response of FcGRIIb-/- mice, a lupus model with an inhibitory-signaling defect, can become exhausted (less subsequent immune-response than the first response) with sequential lipopolysaccharide (LPS) stimulation. Endotoxin tolerance-related modifications of inflammatory response were investigated in FcGRIIb-/- mice in both an in vivo sepsis model and in vitro using cultured macrophages. Serum cytokine concentrations, after the second LPS injection (at 5-fold higher levels than the first dose), did not exceed the first dose levels in either FcGRIIb-/- or wild-type mice. These data indicated an endotoxin-tolerance response in both genetic backgrounds. However, the difference of cytokine levels between the first and second LPS injection was more prominent in FcGRIIb-/- mice. More importantly, CLP-induced sepsis after LPS-preconditioning (two separated doses of LPS administration) was more severe in FcGRIIb-/- mice (as measured by mortality rate, bacteria count in blood, serum cytokines, creatinine, and alanine transaminase). An attenuated response was demonstrated after two sequential LPS stimulations of bone-marrow-derived macrophages. Cytokine production was reduced and lower bacterial killing activity occurred with macrophages from FcGRIIb-/- mice relative to wild-type macrophages. Thus, there is a more prominent effect of endotoxin-tolerance in FcGRIIb-/- macrophages relative to wild-type. In conclusion, repeated-LPS administrations induced quantitatively greater endotoxin-tolerance responses in FcGRIIb-/- mice both in vivo and in vitro. Endotoxin-tolerance in vivo was associated with more severe sepsis, at least in part, due to macrophage-dysfunction.
Collapse
|
48
|
Tan Q, Tai N, Li Y, Pearson J, Pennetti S, Zhou Z, Wong FS, Wen L. Activation-induced cytidine deaminase deficiency accelerates autoimmune diabetes in NOD mice. JCI Insight 2018; 3:95882. [PMID: 29321370 DOI: 10.1172/jci.insight.95882] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 12/05/2017] [Indexed: 01/15/2023] Open
Abstract
B cells play an important role in type 1 diabetes (T1D) development. However, the role of B cell activation-induced cytidine deaminase (AID) in diabetes development is not clear. We hypothesized that AID is important in the immunopathogenesis of T1D. To test this hypothesis, we generated AID-deficient (AID-/-) NOD mice. We found that AID-/-NOD mice developed accelerated T1D, with worse insulitis and high levels of anti-insulin autoantibody in the circulation. Interestingly, neither maternal IgG transferred through placenta, nor IgA transferred through milk affected the accelerated diabetes development. AID-/-NOD mice showed increased activation and proliferation of B and T cells. We found enhanced T-B cell interactions in AID-/-NOD mice, with increased T-bet and IFN-γ expression in CD4+ T cells in the presence of AID-/- B cells. Moreover, excessive lymphoid expansion was observed in AID-/-NOD mice. Importantly, antigen-specific BDC2.5 CD4+ T cells caused more rapid onset of diabetes when cotransferred with AID-/- B cells than when cotransferred with AID+/+ B cells. Thus, our study provides insights into the role of AID in T1D. Our data also suggest that AID is a negative regulator of immune tolerance and ablation of AID can lead to exacerbated islet autoimmunity and accelerated T1D development.
Collapse
Affiliation(s)
- Qiyuan Tan
- Institute of Metabolism and Endocrinology, The Second Xiangya Hospital, Key Laboratory of Diabetes Immunology, Ministry of Education, Central South University, National Clinical Research Center for Metabolic Diseases, Changsha,Hunan, China.,Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Endocrinology and Metabolism, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Ningwen Tai
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yangyang Li
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Endocrinology, Jilin University, Changchun, China
| | - James Pearson
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sean Pennetti
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,School of Medicine, Quinnipiac University, North Haven, Connecticut, USA
| | - Zhiguang Zhou
- Institute of Metabolism and Endocrinology, The Second Xiangya Hospital, Key Laboratory of Diabetes Immunology, Ministry of Education, Central South University, National Clinical Research Center for Metabolic Diseases, Changsha,Hunan, China
| | - F Susan Wong
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Li Wen
- Institute of Metabolism and Endocrinology, The Second Xiangya Hospital, Key Laboratory of Diabetes Immunology, Ministry of Education, Central South University, National Clinical Research Center for Metabolic Diseases, Changsha,Hunan, China.,Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
49
|
Abstract
This update focuses on two main topics. First, recent developments in our understanding of liver sinusoidal endothelial cell (LSEC) function will be reviewed, specifically elimination of blood-borne waste, immunological function of LSECs, interaction of LSECs with liver metastases, LSECs and liver regeneration, and LSECs and hepatic fibrosis. Second, given the current emphasis on rigor and transparency in biomedical research, the update discusses the need for standardization of methods to demonstrate identity and purity of isolated LSECs, pitfalls in methods that might lead to a selection bias in the types of LSECs isolated, and questions about long-term culture of LSECs. Various surface markers used for immunomagnetic selection are reviewed.
Collapse
Affiliation(s)
- Laurie D. DeLeve
- Division of Gastrointestinal and Liver Diseases and the USC Research Center for Liver Diseases, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Ana C. Maretti-Mira
- Division of Gastrointestinal and Liver Diseases and the USC Research Center for Liver Diseases, Keck School of Medicine of the University of Southern California, Los Angeles, California
| |
Collapse
|
50
|
Jhou JP, Chen SJ, Huang HY, Lin WW, Huang DY, Tzeng SJ. Upregulation of FcγRIIB by resveratrol via NF-κB activation reduces B-cell numbers and ameliorates lupus. Exp Mol Med 2017; 49:e381. [PMID: 28960214 PMCID: PMC5628277 DOI: 10.1038/emm.2017.144] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 03/22/2017] [Accepted: 03/29/2017] [Indexed: 12/14/2022] Open
Abstract
Resveratrol, an anti-inflammatory agent, can inhibit pro-inflammatory mediators by activating Sirt1, which is a class III histone deacetylase. However, whether resveratrol can regulate inhibitory or anti-inflammatory molecules has been less studied. FcγRIIB, a receptor for IgG, is an essential inhibitory receptor of B cells for blocking B-cell receptor-mediated activation and for directly inducing apoptosis of B cells. Because mice deficient in either Sirt1 or FcγRIIB develop lupus-like diseases, we investigated whether resveratrol can alleviate lupus through FcγRIIB. We found that resveratrol enhanced the expression of FcγRIIB in B cells, resulting in a marked depletion of plasma cells in the spleen and notably in the bone marrow, thereby decreasing serum autoantibody titers in MRL/lpr mice. The upregulation of FcγRIIB by resveratrol involved an increase of Sirt1 protein and deacetylation of p65 NF-κB (K310). Moreover, increased binding of phosphor-p65 NF-κB (S536) but decreased association of acetylated p65 NF-κB (K310) and phosphor-p65 NF-κB (S468) to the −480 promoter region of Fcgr2b gene was responsible for the resveratrol-mediated enhancement of FcγRIIB gene transcription. Consequently, B cells, especially plasma cells, were considerably reduced in MRL/lpr mice, leading to improvement of nephritis and prolonged survival. Taken together, we provide evidence that pharmacological upregulation of FcγRIIB expression in B cells via resveratrol can selectively reduce B cells, decrease serum autoantibodies and ameliorate lupus nephritis. Our findings lead us to propose FcγRIIB as a new target for therapeutic exploitation, particularly for lupus patients whose FcγRIIB expression levels in B cells are downregulated.
Collapse
Affiliation(s)
- Jyun-Pei Jhou
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Se-Jie Chen
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ho-Yin Huang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wan-Wan Lin
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Duen-Yi Huang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shiang-Jong Tzeng
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|