1
|
Gong Y, Xu R, Gao G, Li S, Liu Y. The role of fatty acid metabolism on B cells and B cell-related autoimmune diseases. Inflamm Res 2025; 74:75. [PMID: 40299047 DOI: 10.1007/s00011-025-02042-3] [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: 02/08/2025] [Revised: 04/08/2025] [Accepted: 04/15/2025] [Indexed: 04/30/2025] Open
Abstract
Fatty acid metabolism plays a critical role in regulating immune cell function, including B cells, which are central to humoral immunity and the pathogenesis of autoimmune diseases. Emerging evidence suggests that fatty acid metabolism influences B cell development, activation, differentiation, and antibody production, thereby impacting B cell-related autoimmune diseases such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and multiple sclerosis (MS). In this review, we discuss the mechanisms by which fatty acid metabolism modulates B cell biology, including energy provision, membrane composition, and signaling pathways. We highlight how alterations in fatty acid synthesis, oxidation, and uptake affect B cell function and contribute to autoimmune pathogenesis. Additionally, we explore the therapeutic potential of targeting fatty acid metabolism in B cells to treat autoimmune diseases. Understanding the interplay between fatty acid metabolism and B cell immunity may provide novel insights into the development of precision therapies for B cell-mediated autoimmune disorders.
Collapse
Affiliation(s)
- Yanmei Gong
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China
| | - Ruiqi Xu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China
| | - Guohui Gao
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China
| | - Simiao Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China
| | - Ying Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Institute of Neuroimmunology, Jinan, Shandong, China.
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China, China.
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, Shandong, China.
| |
Collapse
|
2
|
Togashi T, Ishihara R, Watanabe R, Shiomi M, Yano Y, Fujisawa Y, Katsushima M, Fukumoto K, Yamada S, Hashimoto M. Rheumatoid Factor: Diagnostic and Prognostic Performance and Therapeutic Implications in Rheumatoid Arthritis. J Clin Med 2025; 14:1529. [PMID: 40094988 PMCID: PMC11900400 DOI: 10.3390/jcm14051529] [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: 02/03/2025] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Rheumatoid factor (RF) is the first autoantibody identified in rheumatoid arthritis (RA) which targets the fragment crystallizable (Fc) region of immunoglobulin (Ig) G. Although IgM isotype is predominant, other Ig isotypes, including IgG and IgA, also exist. While RF is not specific to RA, it remains a valuable serological test for diagnosing the disease, as evidenced by its inclusion in the 2010 classification criteria for RA based on elevated serum RF levels. RF is also associated with RA severity, including joint damage and extra-articular manifestations, serving as a poor prognostic factor and aiding in the identification of difficult-to-treat RA. Recent studies have demonstrated that high serum RF levels are associated with a reduced response to tumor necrosis factor (TNF) inhibitors. In contrast, anti-TNF antibodies lacking the Fc portion have shown stable efficacy in RA patients regardless of baseline RF levels. These findings reaffirm the clinical significance of RF measurement, 80 years after its initial discovery. This review explores the diagnostic and prognostic significance of RF and its impact on treatment selection in RA management.
Collapse
Affiliation(s)
| | | | - Ryu Watanabe
- Department of Clinical Immunology, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Arvidsson G, Czarnewski P, Johansson A, Raine A, Imgenberg-Kreuz J, Nordlund J, Nordmark G, Syvänen AC. Multimodal Single-Cell Sequencing of B Cells in Primary Sjögren's Syndrome. Arthritis Rheumatol 2024; 76:255-267. [PMID: 37610265 DOI: 10.1002/art.42683] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/08/2023] [Accepted: 08/18/2023] [Indexed: 08/24/2023]
Abstract
OBJECTIVE B cells are important in the pathogenesis of primary Sjögren's syndrome (pSS). Patients positive for Sjögren's syndrome antigen A/Sjögren syndrome antigen B (SSA/SSB) autoantibodies are more prone to systemic disease manifestations and adverse outcomes. We aimed to determine the role of B cell composition, gene expression, and B cell receptor usage in pSS subgroups stratified for SSA/SSB antibodies. METHODS Over 230,000 B cells were isolated from peripheral blood of patients with pSS (n = 6 SSA-, n = 8 SSA+ single positive and n = 10 SSA/SSB+ double positive) and four healthy controls and processed for single-cell RNA sequencing (scRNA-seq) and single-cell variable, diversity, and joining (VDJ) gene sequencing (scVDJ-seq). RESULTS We show that SSA/SSB+ patients present the highest and lowest proportion of naïve and memory B cells, respectively, and the highest up-regulation of interferon-induced genes across all B cell subtypes. Differential usage of IGHV showed that IGHV1-69 and IGHV4-30-4 were more often used in all pSS subgroups compared with controls. Memory B cells from SSA/SSB+ patients displayed a higher proportion of cells with unmutated VDJ transcripts compared with other pSS patient groups and controls, indicating altered somatic hypermutation processes. Comparison with previous studies revealed heterogeneous clonotype pools, with little overlap in CDR3 sequences. Joint analysis using scRNA-seq and scVDJ-seq data allowed unsupervised stratification of patients with pSS and identified novel parameters that correlated to disease manifestations and antibody status. CONCLUSION We describe heterogeneity and molecular characteristics in B cells from patients with pSS, providing clues to intrinsic differences in B cells that affect the phenotype and outcome and allowing stratification of patients with pSS at improved resolution.
Collapse
|
4
|
Lee AYS, Wang JJ, Gordon TP, Reed JH. Phases and Natural History of Sjögren's Disease: A New Model for an Old Disease? Arthritis Care Res (Hoboken) 2023; 75:1580-1587. [PMID: 36063396 PMCID: PMC10953327 DOI: 10.1002/acr.25011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/23/2022] [Accepted: 08/30/2022] [Indexed: 01/07/2023]
Abstract
Sjögren's disease (SjD) is an archetypal and heterogenous autoimmune disorder that is characterized by exocrine glandular dysfunction. A proportion of patients develop severe extraglandular manifestations, such as cryoglobulinemia, and have an increased risk of lymphoma, both of which can adversely affect quality of life and occasionally mortality. As with most autoimmune disorders, the pathogenesis is poorly understood and difficult to predict, and, frustratingly, there is a lack of targeted therapies to cure this disease. We review the disease manifestations of SjD and propose a staged model for understanding the evolution of pathology. In longitudinal studies, most patients remain relatively stable in terms of their laboratory and clinical parameters. However, in the setting of various risk factors, a proportion of patients develop severe symptoms and/or lymphoma. We discuss potential underlying mechanisms for disease progression and the strengths and limitations of using a staged model to correlate the pathogenesis and spectrum of manifestations in SjD. Ultimately, understanding how and why some patients remain relatively stable, whereas others progress and develop florid systemic disease and a fraction develop lymphoma, is key to developing preventative and therapeutic treatments.
Collapse
Affiliation(s)
- Adrian Y. S. Lee
- The Westmead Institute for Medical ResearchUniversity of Sydney, Westmead Hospital, NSW Health PathologyWestmeadNew South WalesAustralia
| | - Jing Jing Wang
- SA Pathology and Flinders UniversityBedford ParkSouth AustraliaAustralia
| | - Tom P. Gordon
- SA Pathology and Flinders UniversityBedford ParkSouth AustraliaAustralia
| | - Joanne H. Reed
- The Westmead Institute for Medical ResearchUniversity of SydneyWestmeadNew South WalesAustralia
| |
Collapse
|
5
|
Troelnikov A, Armour B, Putty T, Aggarwal A, Akerman A, Milogiannakis V, Chataway T, King J, Turville SG, Gordon TP, Wang JJ. Immunoglobulin repertoire restriction characterizes the serological responses of patients with predominantly antibody deficiency. J Allergy Clin Immunol 2023; 152:290-301.e7. [PMID: 36965845 DOI: 10.1016/j.jaci.2023.02.033] [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: 08/22/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 03/27/2023]
Abstract
BACKGROUND Predominantly antibody deficiency (PAD) is the most common category of inborn errors of immunity and is underpinned by impaired generation of appropriate antibody diversity and quantity. In the clinic, responses are interrogated by assessment of vaccination responses, which is central to many PAD diagnoses. However, the composition of the generated antibody repertoire is concealed from traditional quantitative measures of serological responses. Leveraging modern mass spectrometry-based proteomics (MS-proteomics), it is possible to elaborate the molecular features of specific antibody repertoires, which may address current limitations of diagnostic vaccinology. OBJECTIVES We sought to evaluate serum antibody responses in patients with PAD following vaccination with a neo-antigen (severe acute respiratory syndrome coronavirus-2 vaccination) using MS-proteomics. METHODS Following severe acute respiratory syndrome coronavirus-2 vaccination, serological responses in individuals with PAD and healthy controls (HCs) were assessed by anti-S1 subunit ELISA and neutralization assays. Purified anti-S1 subunit IgG and IgM was profiled by MS-proteomics for IGHV subfamily usage and somatic hypermutation analysis. RESULTS Twelve patients with PAD who were vaccine-responsive were recruited with 11 matched vaccinated HCs. Neutralization and end point anti-S1 titers were lower in PAD. All subjects with PAD demonstrated restricted anti-S1 IgG antibody repertoires, with usage of <5 IGHV subfamilies (median: 3; range 2-4), compared to ≥5 for the 11 HC subjects (P < .001). IGHV3-7 utilization was far less common in patients with PAD than in HCs (2 of 12 vs 10 of 11; P = .001). Amino acid substitutions due to somatic hypermutation per subfamily did not differ between groups. Anti-S1 IgM was present in 64% and 50% of HC and PAD cohorts, respectively, and did not differ significantly between HCs and patients with PAD. CONCLUSIONS This study demonstrates the breadth of anti-S1 antibodies elicited by vaccination at the proteome level and identifies stereotypical restriction of IGHV utilization in the IgG repertoire in patients with PAD compared with HC subjects. Despite uniformly pauci-clonal antibody repertoires some patients with PAD generated potent serological responses, highlighting a possible limitation of traditional serological techniques. These findings suggest that IgG repertoire restriction is a key feature of antibody repertoires in PAD.
Collapse
Affiliation(s)
- Alexander Troelnikov
- College of Medicine and Public Health, Flinders University, Bedford Park, Australia; SA Pathology, Adelaide, Australia.
| | - Bridie Armour
- College of Medicine and Public Health, Flinders University, Bedford Park, Australia; SA Pathology, Adelaide, Australia
| | - Trishni Putty
- College of Medicine and Public Health, Flinders University, Bedford Park, Australia; SA Pathology, Adelaide, Australia
| | | | | | | | - Tim Chataway
- College of Medicine and Public Health, Flinders University, Bedford Park, Australia
| | - Jovanka King
- SA Pathology, Adelaide, Australia; Women's and Children's Hospital Network, Adelaide, Australia; Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | | | - Tom P Gordon
- College of Medicine and Public Health, Flinders University, Bedford Park, Australia; SA Pathology, Adelaide, Australia; Flinders Medical Centre, Bedford Park, Australia
| | - Jing Jing Wang
- College of Medicine and Public Health, Flinders University, Bedford Park, Australia; SA Pathology, Adelaide, Australia
| |
Collapse
|
6
|
Kamounah S, Sembler-Møller ML, Nielsen CH, Pedersen AML. Sjögren's syndrome: novel insights from proteomics and miRNA expression analysis. Front Immunol 2023; 14:1183195. [PMID: 37275849 PMCID: PMC10232878 DOI: 10.3389/fimmu.2023.1183195] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/09/2023] [Indexed: 06/07/2023] Open
Abstract
Introduction Sjögren's syndrome (SS) is a systemic autoimmune disease, which affects the exocrine glands leading to glandular dysfunction and, particularly, symptoms of oral and ocular dryness. The aetiology of SS remains unclear, and the disease lacks distinctive clinical features. The current diagnostic work-up is complex, invasive and often time-consuming. Thus, there is an emerging need for identifying disease-specific and, ideally, non-invasive immunological and molecular biomarkers that can simplify the diagnostic process, allow stratification of patients, and assist in monitoring the disease course and outcome of therapeutic intervention in SS. Methods This systematic review addresses the use of proteomics and miRNA-expression profile analyses in this regard. Results and discussion Out of 272 papers that were identified and 108 reviewed, a total of 42 papers on proteomics and 23 papers on miRNA analyses in saliva, blood and salivary gland tissue were included in this review. Overall, the proteomic and miRNA studies revealed considerable variations with regard to candidate biomarker proteins and miRNAs, most likely due to variation in sample size, processing and analytical methods, but also reflecting the complexity of SS and patient heterogeneity. However, interesting novel knowledge has emerged and further validation is needed to confirm their potential role as biomarkers in SS.
Collapse
Affiliation(s)
- Sarah Kamounah
- Section for Oral Biology and Immunopathology/Oral Medicine, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maria Lynn Sembler-Møller
- Section for Oral Biology and Immunopathology/Oral Medicine, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claus Henrik Nielsen
- Section for Oral Biology and Immunopathology/Oral Medicine, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Rheumatology and Spine Diseases, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Anne Marie Lynge Pedersen
- Section for Oral Biology and Immunopathology/Oral Medicine, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Del Padre M, Marrapodi R, Minafò YA, Piano Mortari E, Radicchio G, Bocci C, Gragnani L, Camponeschi A, Colantuono S, Stefanini L, Basili S, Carsetti R, Fiorilli M, Casato M, Visentini M. Dual stimulation by autoantigen and CpG fosters the proliferation of exhausted rheumatoid factor-specific CD21 low B cells in hepatitis C virus-cured mixed cryoglobulinemia. Front Immunol 2023; 14:1094871. [PMID: 36845129 PMCID: PMC9945227 DOI: 10.3389/fimmu.2023.1094871] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/20/2023] [Indexed: 02/10/2023] Open
Abstract
Introduction Hepatitis C virus (HCV) causes mixed cryoglobulinemia (MC) by driving clonal expansion of B cells expressing B cell receptors (BCRs), often encoded by the VH1-69 variable gene, endowed with both rheumatoid factor (RF) and anti-HCV specificity. These cells display an atypical CD21low phenotype and functional exhaustion evidenced by unresponsiveness to BCR and Toll-like receptor 9 (TLR9) stimuli. Although antiviral therapy is effective on MC vasculitis, pathogenic B cell clones persist long thereafter and can cause virus-independent disease relapses. Methods Clonal B cells from patients with HCV-associated type 2 MC or healthy donors were stimulated with CpG or heath-aggregated IgG (as surrogate immune complexes) alone or in combination; proliferation and differentiation were then evaluated by flow cytometry. Phosphorylation of AKT and of the p65 NF-kB subunit were measured by flow cytometry. TLR9 was quantified by qPCR and by intracellular flow cytometry, and MyD88 isoforms were analyzed using RT-PCR. Discussion We found that dual triggering with autoantigen and CpG restored the capacity of exhausted VH1-69pos B cells to proliferate. The signaling mechanism for this BCR/TLR9 crosstalk remains elusive, since TLR9 mRNA and protein as well as MyD88 mRNA were normally expressed and CpG-induced phosphorylation of p65 NF-kB was intact in MC clonal B cells, whereas BCR-induced p65 NF-kB phosphorylation was impaired and PI3K/Akt signaling was intact. Our findings indicate that autoantigen and CpG of microbial or cellular origin may unite to foster persistence of pathogenic RF B cells in HCV-cured MC patients. BCR/TLR9 crosstalk might represent a more general mechanism enhancing systemic autoimmunity by the rescue of exhausted autoreactive CD21low B cells.
Collapse
Affiliation(s)
- Martina Del Padre
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Ramona Marrapodi
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Ylenia A Minafò
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Eva Piano Mortari
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- B cell unit, Immunology Research Area, IRCCS Bambino Gesù Children's Hospital, Florence, Italy
| | - Giovanna Radicchio
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Chiara Bocci
- B cell unit, Immunology Research Area, IRCCS Bambino Gesù Children's Hospital, Florence, Italy
| | - Laura Gragnani
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandro Camponeschi
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Stefania Colantuono
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucia Stefanini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Stefania Basili
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Rita Carsetti
- B cell unit, Immunology Research Area, IRCCS Bambino Gesù Children's Hospital, Florence, Italy
| | - Massimo Fiorilli
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Milvia Casato
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Marcella Visentini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
8
|
Wang JJ, Armour B, Chataway T, Troelnikov A, Colella A, Yacoub O, Hockley S, Tan CW, Gordon TP. Vaccine-induced immune thrombotic thrombocytopenia is mediated by a stereotyped clonotypic antibody. Blood 2022; 140:1738-1742. [PMID: 35661872 PMCID: PMC9906116 DOI: 10.1182/blood.2022016474] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/31/2022] [Indexed: 11/20/2022] Open
Affiliation(s)
- Jing Jing Wang
- Department of Immunology, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
- Department of Immunology, South Australia (SA) Pathology (Flinders Medical Centre), Bedford Park, SA, Australia
| | - Bridie Armour
- Department of Immunology, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
- Department of Immunology, South Australia (SA) Pathology (Flinders Medical Centre), Bedford Park, SA, Australia
| | - Tim Chataway
- Flinders Proteomics Facility, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Alexander Troelnikov
- Department of Immunology, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
- Department of Immunology, South Australia (SA) Pathology (Flinders Medical Centre), Bedford Park, SA, Australia
| | - Alex Colella
- Flinders Proteomics Facility, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | | | - Simon Hockley
- Intensive Care Unit, Calvary Hospital, Adelaide, SA, Australia
| | - Chee Wee Tan
- SA Pathology, Adelaide, SA, Australia
- Department of Haematology, Royal Adelaide Hospital, Central Area Local Health Network (CALHN), Adelaide, SA, Australia
- Department of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Tom Paul Gordon
- Department of Immunology, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
- Department of Immunology, South Australia (SA) Pathology (Flinders Medical Centre), Bedford Park, SA, Australia
| |
Collapse
|
9
|
Liu P, Wu J, Sun D, Li H, Qi Z, Tang X, Su W, Li Y, Qin X. Proteomic Profiling of Cryoglobulinemia. Front Immunol 2022; 13:855513. [PMID: 35677050 PMCID: PMC9167934 DOI: 10.3389/fimmu.2022.855513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Objective We aimed to explore and identify candidate protein biomarkers of cryoglobulinemia (CGE) in disease control patients with negative cryoglobulin (DC) or healthy controls (HCs). Methods The tandem mass tag (TMT)-labeled serum quantitative proteomics approach was used to identify differentially expressed proteins between the CGE and DC groups. Ingenuity pathway analysis was used for functional annotation of differentially expressed proteins. Biomarker candidates were validated in another cohort using the parallel reaction monitoring (PRM) method. Apolipoprotein A1 (APOA1), apolipoprotein CIII (APOC3), adiponectin, and proprotein convertase subtilisin/kexin type-9 (PCSK9), which represent key proteins involved in the cholesterol metabolism pathway, were further verified in an increased number of samples by enzyme-linked immunosorbent assay (ELISA). Results A total of 1004 proteins were identified, of which 109 proteins were differentially expressed between the CGE and DC groups. These differentially expressed proteins were primarily involved in hepatic fibrosis/hepatic stellate cell activation and immune/inflammation-related pathways. In the disease and biofunction analysis, these proteins were mainly associated with the adhesion of blood cells, leukocyte migration, cholesterol transport, and transport of lipids. Twelve candidate biomarkers were validated by PRM-based proteomics, and proteins involved in the cholesterol metabolism pathway were further verified. APOA1, APOC3, adiponectin and PCSK9 concentrations were increased in CGE patients compared with healthy controls (P=0.0123, 0.1136, 0.5760, and 0.0019, respectively). Conclusion This report describes the first application of a TMT-PRM-ELISA workflow to identify and validate CGE-specific biomarkers in serum. APOA1 and PCSK9 have been confirmed to be increased in CGE patients, demonstrating that proteins involved in cholesterol metabolism are also implicated in the development of CGE. These findings contribute to pathogenesis research and biomarker discovery in CGE.
Collapse
Affiliation(s)
- Peng Liu
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianqiang Wu
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dandan Sun
- Department of Laboratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China
| | - Haolong Li
- Department of Laboratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China
| | - Zhihong Qi
- Department of Laboratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China
| | - Xiaoyue Tang
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Su
- Department of Laboratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Xuzhen Qin, ; Wei Su, ; Yongzhe Li,
| | - Yongzhe Li
- Department of Laboratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Xuzhen Qin, ; Wei Su, ; Yongzhe Li,
| | - Xuzhen Qin
- Department of Laboratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Xuzhen Qin, ; Wei Su, ; Yongzhe Li,
| |
Collapse
|
10
|
Reed JH. Transforming mutations in the development of pathogenic B cell clones and autoantibodies. Immunol Rev 2022; 307:101-115. [PMID: 35001403 DOI: 10.1111/imr.13064] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/31/2021] [Accepted: 01/01/2022] [Indexed: 12/16/2022]
Abstract
Autoimmune diseases are characterized by serum autoantibodies, some of which are pathogenic, causing severe manifestations and organ injury. However, autoantibodies of the same antigenic reactivity are also present in the serum of asymptomatic people years before they develop any clinical signs of autoimmunity. Autoantibodies can arise during multiple stages of B cell development, and various genetic and environmental factors drive their production. However, what drives the development of pathogenic autoantibodies is poorly understood. Advances in single-cell technology have enabled the deep analysis of rare B cell clones producing pathogenic autoantibodies responsible for vasculitis in patients with primary Sjögren's syndrome complicated by mixed cryoglobulinaemia. These findings demonstrated a cascade of genetic events involving stereotypic immunoglobulin V(D)J recombination and transforming somatic mutations in lymphoma genes and V(D)J regions that disrupted antibody quality control mechanisms and decreased autoantibody solubility. Most studies consider V(D)J mutations that enhance autoantibody affinity to drive pathology; however, V(D)J mutations that increase autoantibody propensity to form insoluble complexes could be a major contributor to autoantibody pathogenicity. Defining the molecular characteristics of pathogenic autoantibodies and failed tolerance checkpoints driving their formation will improve prognostication, enabling early treatment to prevent escalating organ damage and B cell malignancy.
Collapse
Affiliation(s)
- Joanne H Reed
- Westmead Institute for Medical Research, Centre for Immunology and Allergy Research, Westmead, NSW, Australia.,Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
11
|
Wheatley AK, Juno JA, Wang JJ, Selva KJ, Reynaldi A, Tan HX, Lee WS, Wragg KM, Kelly HG, Esterbauer R, Davis SK, Kent HE, Mordant FL, Schlub TE, Gordon DL, Khoury DS, Subbarao K, Cromer D, Gordon TP, Chung AW, Davenport MP, Kent SJ. Evolution of immune responses to SARS-CoV-2 in mild-moderate COVID-19. Nat Commun 2021; 12:1162. [PMID: 33608522 PMCID: PMC7896046 DOI: 10.1038/s41467-021-21444-5] [Citation(s) in RCA: 255] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 01/19/2021] [Indexed: 01/05/2023] Open
Abstract
The durability of infection-induced SARS-CoV-2 immunity has major implications for reinfection and vaccine development. Here, we show a comprehensive profile of antibody, B cell and T cell dynamics over time in a cohort of patients who have recovered from mild-moderate COVID-19. Binding and neutralising antibody responses, together with individual serum clonotypes, decay over the first 4 months post-infection. A similar decline in Spike-specific CD4+ and circulating T follicular helper frequencies occurs. By contrast, S-specific IgG+ memory B cells consistently accumulate over time, eventually comprising a substantial fraction of circulating the memory B cell pool. Modelling of the concomitant immune kinetics predicts maintenance of serological neutralising activity above a titre of 1:40 in 50% of convalescent participants to 74 days, although there is probably additive protection from B cell and T cell immunity. This study indicates that SARS-CoV-2 immunity after infection might be transiently protective at a population level. Therefore, SARS-CoV-2 vaccines might require greater immunogenicity and durability than natural infection to drive long-term protection.
Collapse
Affiliation(s)
- Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Australian Research Council Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, VIC, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Jing J Wang
- Department of Immunology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Kevin J Selva
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Wen Shi Lee
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Kathleen M Wragg
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Hannah G Kelly
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Australian Research Council Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, VIC, Australia
| | - Robyn Esterbauer
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Australian Research Council Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, VIC, Australia
| | - Samantha K Davis
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Helen E Kent
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Francesca L Mordant
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Timothy E Schlub
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - David L Gordon
- Department of Microbiology and Infectious Diseases, Flinders University and SA Pathology, Flinders Medical Centre, Adelaide, SA, Australia
| | - David S Khoury
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Deborah Cromer
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Tom P Gordon
- Department of Immunology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Immunology, SA Pathology, Flinders Medical Centre, Adelaide, SA, Australia
| | - Amy W Chung
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Miles P Davenport
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
- Australian Research Council Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, VIC, Australia.
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
12
|
Wang JJ, Lee AY, Colella AD, Chataway TK, Gordon TP, Wechalekar MD. Proteomic mapping of rheumatoid factors in early rheumatoid arthritis. Arthritis Rheumatol 2020; 72:2159-2161. [DOI: 10.1002/art.41446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 06/27/2020] [Indexed: 12/27/2022]
Affiliation(s)
- Jing J. Wang
- Flinders Medical Centre and Flinders University Bedford Park South Australia Australia
| | - Adrian Y.S. Lee
- Flinders Medical Centre and Flinders University Bedford Park South Australia Australia
| | - Alex D. Colella
- Flinders Medical Centre and Flinders University Bedford Park South Australia Australia
| | - Tim K. Chataway
- Flinders Medical Centre and Flinders University Bedford Park South Australia Australia
| | - Tom P. Gordon
- Flinders Medical Centre and Flinders University Bedford Park South Australia Australia
| | - Mihir D. Wechalekar
- Flinders Medical Centre and Flinders University Bedford Park South Australia Australia
| |
Collapse
|
13
|
Lau A, Avery DT, Jackson K, Lenthall H, Volpi S, Brigden H, Russell AJ, Bier J, Reed JH, Smart JM, Cole T, Choo S, Gray PE, Berglund LJ, Hsu P, Wong M, O'Sullivan M, Boztug K, Meyts I, Uzel G, Notarangelo LD, Brink R, Goodnow CC, Tangye SG, Deenick EK. Activated PI3Kδ breaches multiple B cell tolerance checkpoints and causes autoantibody production. J Exp Med 2020; 217:132760. [PMID: 31841125 PMCID: PMC7041712 DOI: 10.1084/jem.20191336] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/29/2019] [Accepted: 11/07/2019] [Indexed: 12/17/2022] Open
Abstract
In patients, gain-of-function (GOF) mutations in PIK3CD break tolerance, causing highly penetrant secretion of autoreactive IgM. Mouse models reveal that Pik3cd GOF subverts the response to self-antigen, preventing the induction of anergy and instead stimulating plasmablast and GC formation. Antibody-mediated autoimmune diseases are a major health burden. However, our understanding of how self-reactive B cells escape self-tolerance checkpoints to secrete pathogenic autoantibodies remains incomplete. Here, we demonstrate that patients with monogenic immune dysregulation caused by gain-of-function mutations in PIK3CD, encoding the p110δ catalytic subunit of phosphoinositide 3-kinase (PI3K), have highly penetrant secretion of autoreactive IgM antibodies. In mice with the corresponding heterozygous Pik3cd activating mutation, self-reactive B cells exhibit a cell-autonomous subversion of their response to self-antigen: instead of becoming tolerized and repressed from secreting autoantibody, Pik3cd gain-of-function B cells are activated by self-antigen to form plasmablasts that secrete high titers of germline-encoded IgM autoantibody and hypermutating germinal center B cells. However, within the germinal center, peripheral tolerance was still enforced, and there was selection against B cells with high affinity for self-antigen. These data show that the strength of PI3K signaling is a key regulator of pregerminal center B cell self-tolerance and thus represents a druggable pathway to treat antibody-mediated autoimmunity.
Collapse
Affiliation(s)
- Anthony Lau
- Immunity and Inflammatory Diseases, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.,St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Danielle T Avery
- Immunity and Inflammatory Diseases, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Katherine Jackson
- Immunity and Inflammatory Diseases, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Helen Lenthall
- Immunity and Inflammatory Diseases, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Stefano Volpi
- Clinica Pediatrica e Reumatologia, Centro per le malattie Autoinfiammatorie e Immunodeficienze, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini and Dipartimento di Neuroscienze, riabilitazione, oftalmologia, genetica e scienze materno-infantili (DINOGMI), Università degli Studi di Genova, Genova, Italy
| | - Henry Brigden
- Immunity and Inflammatory Diseases, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Amanda J Russell
- Immunity and Inflammatory Diseases, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Julia Bier
- Immunity and Inflammatory Diseases, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.,St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Joanne H Reed
- Immunity and Inflammatory Diseases, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.,St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
| | - Joanne M Smart
- Department of Allergy and Immunology, Royal Children's Hospital Melbourne, Victoria, Australia
| | - Theresa Cole
- Department of Allergy and Immunology, Royal Children's Hospital Melbourne, Victoria, Australia
| | - Sharon Choo
- Department of Allergy and Immunology, Royal Children's Hospital Melbourne, Victoria, Australia
| | - Paul E Gray
- School of Women's and Children's Health, UNSW Sydney, Sydney, Australia.,Clinical Immunogenomics Research Consortium of Australasia, Sydney, Australia
| | - Lucinda J Berglund
- Clinical Immunogenomics Research Consortium of Australasia, Sydney, Australia.,Immunopathology Department, Westmead Hospital, Westmead, New South Wales, Australia.,Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Peter Hsu
- Clinical Immunogenomics Research Consortium of Australasia, Sydney, Australia.,Children's Hospital at Westmead, New South Wales, Australia
| | - Melanie Wong
- Clinical Immunogenomics Research Consortium of Australasia, Sydney, Australia.,Children's Hospital at Westmead, New South Wales, Australia
| | - Michael O'Sullivan
- Clinical Immunogenomics Research Consortium of Australasia, Sydney, Australia.,Department of Immunology and Allergy, Princess Margaret Hospital, Subiaco, Western Australia, Australia
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,St. Anna Children's Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Isabelle Meyts
- Department of Immunology and Microbiology, Inborn Errors of Immunity, Department of Pediatrics, University Hospitals Leuven and KU Leuven, Leuven, Belgium
| | - Gulbu Uzel
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Robert Brink
- Immunity and Inflammatory Diseases, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.,St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia.,Clinical Immunogenomics Research Consortium of Australasia, Sydney, Australia
| | - Christopher C Goodnow
- Immunity and Inflammatory Diseases, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.,Clinical Immunogenomics Research Consortium of Australasia, Sydney, Australia.,UNSW Cellular Genomics Futures Institute, UNSW Sydney, Sydney, Australia
| | - Stuart G Tangye
- Immunity and Inflammatory Diseases, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.,St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia.,Clinical Immunogenomics Research Consortium of Australasia, Sydney, Australia
| | - Elissa K Deenick
- Immunity and Inflammatory Diseases, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.,Clinical Immunogenomics Research Consortium of Australasia, Sydney, Australia.,Faculty of Medicine, UNSW Sydney, Sydney, Australia
| |
Collapse
|
14
|
Falkenburg WJJ, Oskam N, Koers J, van Boheemen L, Ooijevaar-de Heer P, Verstappen GM, Bootsma H, Kroese FGM, van Schaardenburg D, Wolbink G, Rispens T. Identification of Clinically and Pathophysiologically Relevant Rheumatoid Factor Epitopes by Engineered IgG Targets. Arthritis Rheumatol 2020; 72:2005-2016. [PMID: 32648642 DOI: 10.1002/art.41430] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 06/16/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Rheumatoid factors (RFs), which are anti-IgG autoantibodies strongly associated with rheumatoid arthritis (RA), are also found in other diseases and in healthy individuals. RFs bind to various epitopes in the constant (Fc-) domain of IgG. Therefore, disease-specific reactivity patterns may exist. This study was undertaken in order to develop a new approach to dissecting RF epitope binding patterns across different diseases. METHODS We analyzed RF reactivity patterns in serum from patients with seropositive arthralgia, patients with RA, and patients with primary Sjögren's syndrome (SS) using bioengineered, natively folded IgG-Fc targets that demonstrated selective RF binding toward several distinct regions of the IgG-Fc domain. RESULTS Rheumatoid factor responses primarily bound the Fc Elbow region, with a smaller number of RFs binding the Fc Tail region, while the Fc receptor binding region was hardly targeted. A restricted reactivity against the IgG-Fc Tail region was associated with less positivity for anti-citrullinated protein antibodies (ACPAs) and less arthritis development in arthralgia, whereas combined reactivity toward IgG-Fc Tail and Elbow regions was associated with more arthritis development. Reactivity toward the IgG-Fc Tail region was observed far more frequently in RA than in primary SS. CONCLUSION Bioengineered IgG targets enable serologic characterization of RF reactivity patterns, and use of this approach appears to reveal patterns associated with ACPA detection and arthritis development in patients with arthralgia. These patterns are able to distinguish RA patients from primary SS patients. This new methodology improves the clinical value of RFs and our understanding of their pathophysiologic processes.
Collapse
Affiliation(s)
- Willem J J Falkenburg
- Amsterdam Rheumatology and Immunology Center, Reade, Sanquin Research, and Academic Medical Center, Amsterdam, The Netherlands
| | - Nienke Oskam
- Sanquin Research and Academic Medical Center, Amsterdam, The Netherlands
| | - Jana Koers
- Sanquin Research and Academic Medical Center, Amsterdam, The Netherlands
| | | | | | - Gwenny M Verstappen
- University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Hendrika Bootsma
- University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Frans G M Kroese
- University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Dirkjan van Schaardenburg
- Amsterdam Rheumatology and Immunology Center, Reade, and, Academic Medical Center, Amsterdam, The Netherlands
| | - Gertjan Wolbink
- Amsterdam Rheumatology and Immunology Center and Reade, Amsterdam, The Netherlands
| | - Theo Rispens
- Sanquin Research and Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Bende RJ, Janssen J, Beentjes A, Wormhoudt TAM, Wagner K, Haacke EA, Kroese FGM, Guikema JEJ, van Noesel CJM. Salivary Gland Mucosa-Associated Lymphoid Tissue-Type Lymphoma From Sjögren's Syndrome Patients in the Majority Express Rheumatoid Factors Affinity-Selected for IgG. Arthritis Rheumatol 2020; 72:1330-1340. [PMID: 32182401 PMCID: PMC7496822 DOI: 10.1002/art.41263] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/12/2020] [Indexed: 12/13/2022]
Abstract
Objective Patients with Sjӧgren's syndrome (SS) have an increased risk of developing malignant B cell lymphomas, particularly mucosa‐associated lymphoid tissue (MALT)–type lymphomas. We have previously shown that a predominant proportion of patients with SS‐associated salivary gland MALT lymphoma express somatically hypermutated IgM with strong amino acid sequence homology with stereotypic rheumatoid factors (RFs). The present study was undertaken in a larger cohort of patients with SS‐associated MALT lymphoma to more firmly assess the frequency of RF reactivity and the significance of somatic IGV‐region mutations for RF reactivity. Methods B cell antigen receptors (BCRs) of 16 patients with SS‐associated salivary gland MALT lymphoma were analyzed. Soluble recombinant IgM was produced of 12 MALT lymphoma samples, including 1 MALT lymphoma sample that expressed an IgM antibody fitting in a novel IGHV3‐30–encoded stereotypic IGHV subset. For 4 of the 12 IgM antibodies from MALT lymphoma samples, the somatically mutated IGHV and IGKV gene sequences were reverted to germline configurations. Their RF activity and binding affinity were determined by enzyme‐linked immunosorbent assay and surface plasmon resonance, respectively. Results Nine (75%) of the 12 IgM antibodies identified in patients with SS‐associated salivary gland MALT lymphoma displayed strong monoreactive RF activity. Reversion of the IGHV and IGKV mutations to germline configuration resulted in RF affinities for IgG that were significantly lower for 3 of the 4 somatically mutated IgM antibodies. In stereotypic IGHV3‐7/IGKV3‐15–encoded RFs, a recurrent replacement mutation in the IGKV3‐15–third complementarity‐determining region was found to play a pivotal role in the affinity for IgG‐Fc. Conclusion A majority of patients with SS‐associated salivary gland MALT lymphoma express somatically mutated BCRs that are selected for monoreactive, high‐affinity binding of IgG‐Fc. These data underscore the notion that soluble IgG, most likely in immune complexes in inflamed tissues, is the principal autoantigen in the pathogenesis of a variety of B cell lymphomas, particularly SS‐associated MALT lymphomas.
Collapse
Affiliation(s)
- Richard J Bende
- Amsterdam University Medical Center and University of Amsterdam, Amsterdam, The Netherlands
| | - Jerry Janssen
- Amsterdam University Medical Center and University of Amsterdam, Amsterdam, The Netherlands
| | - Anna Beentjes
- Amsterdam University Medical Center and University of Amsterdam, Amsterdam, The Netherlands
| | - Thera A M Wormhoudt
- Amsterdam University Medical Center and University of Amsterdam, Amsterdam, The Netherlands
| | - Koen Wagner
- AIMM Therapeutics, Amsterdam, The Netherlands
| | - Erlin A Haacke
- University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Frans G M Kroese
- University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Jeroen E J Guikema
- Amsterdam University Medical Center and University of Amsterdam, Amsterdam, The Netherlands
| | - Carel J M van Noesel
- Amsterdam University Medical Center and University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Singh M, Jackson KJL, Wang JJ, Schofield P, Field MA, Koppstein D, Peters TJ, Burnett DL, Rizzetto S, Nevoltris D, Masle-Farquhar E, Faulks ML, Russell A, Gokal D, Hanioka A, Horikawa K, Colella AD, Chataway TK, Blackburn J, Mercer TR, Langley DB, Goodall DM, Jefferis R, Gangadharan Komala M, Kelleher AD, Suan D, Rischmueller M, Christ D, Brink R, Luciani F, Gordon TP, Goodnow CC, Reed JH. Lymphoma Driver Mutations in the Pathogenic Evolution of an Iconic Human Autoantibody. Cell 2020; 180:878-894.e19. [PMID: 32059783 DOI: 10.1016/j.cell.2020.01.029] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 11/11/2019] [Accepted: 01/22/2020] [Indexed: 12/26/2022]
Abstract
Pathogenic autoantibodies arise in many autoimmune diseases, but it is not understood how the cells making them evade immune checkpoints. Here, single-cell multi-omics analysis demonstrates a shared mechanism with lymphoid malignancy in the formation of public rheumatoid factor autoantibodies responsible for mixed cryoglobulinemic vasculitis. By combining single-cell DNA and RNA sequencing with serum antibody peptide sequencing and antibody synthesis, rare circulating B lymphocytes making pathogenic autoantibodies were found to comprise clonal trees accumulating mutations. Lymphoma driver mutations in genes regulating B cell proliferation and V(D)J mutation (CARD11, TNFAIP3, CCND3, ID3, BTG2, and KLHL6) were present in rogue B cells producing the pathogenic autoantibody. Antibody V(D)J mutations conferred pathogenicity by causing the antigen-bound autoantibodies to undergo phase transition to insoluble aggregates at lower temperatures. These results reveal a pre-neoplastic stage in human lymphomagenesis and a cascade of somatic mutations leading to an iconic pathogenic autoantibody.
Collapse
Affiliation(s)
- Mandeep Singh
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | | | - Jing J Wang
- Department of Immunology, Flinders University and SA Pathology, Bedford Park, SA 5042, Australia
| | - Peter Schofield
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Matt A Field
- Australian Institute of Tropical Health and Medicine and Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Smithfield, QLD 4878, Australia; The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
| | - David Koppstein
- Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Timothy J Peters
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Deborah L Burnett
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Simone Rizzetto
- Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Damien Nevoltris
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Etienne Masle-Farquhar
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Megan L Faulks
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Amanda Russell
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Divya Gokal
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Asami Hanioka
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia; Tokyo Medical and Dental University, Tokyo 113-851, Japan
| | - Keisuke Horikawa
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
| | - Alexander D Colella
- Department of Immunology, Flinders University and SA Pathology, Bedford Park, SA 5042, Australia; Flinders Proteomics Facility, Flinders University, Bedford Park, SA 5042, Australia
| | - Timothy K Chataway
- Flinders Proteomics Facility, Flinders University, Bedford Park, SA 5042, Australia
| | - James Blackburn
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Tim R Mercer
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia; Altius Institute for Biomedical Sciences, Seattle, WA 98121, USA
| | - David B Langley
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - D Margaret Goodall
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Roy Jefferis
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | | | - Anthony D Kelleher
- Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Dan Suan
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; Westmead Clinical School, The University of Sydney, Westmead, NSW 2145, Australia
| | - Maureen Rischmueller
- Rheumatology Department, The Queen Elizabeth Hospital and Discipline of Medicine, University of Adelaide, Woodville South, SA 5011, Australia
| | - Daniel Christ
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Robert Brink
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Fabio Luciani
- Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, NSW 2052, Australia; School of Medical Sciences and Cellular Genomics Futures Institute, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Tom P Gordon
- Department of Immunology, Flinders University and SA Pathology, Bedford Park, SA 5042, Australia
| | - Christopher C Goodnow
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; School of Medical Sciences and Cellular Genomics Futures Institute, UNSW Sydney, Sydney, NSW 2052, Australia.
| | - Joanne H Reed
- The Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia.
| |
Collapse
|
17
|
Lee AY, Chataway T, Gordon TP, Wang JJ. Molecular typing of cryoglobulins by mass spectrometry. Ann Rheum Dis 2020; 79:163-164. [PMID: 31653649 DOI: 10.1136/annrheumdis-2019-216091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 10/13/2019] [Indexed: 11/04/2022]
Affiliation(s)
- Adrian Ys Lee
- Department of Immunology, SA Pathology (Flinders Medical Centre), Bedford Park, SA, Australia
- Department of Immunology, Flinders University, Bedford Park, SA, Australia
| | - Tim Chataway
- Flinders Proteomics Facility, Flinders University, Bedford Park, SA, Australia
| | - Thomas P Gordon
- Department of Immunology, SA Pathology (Flinders Medical Centre), Bedford Park, SA, Australia
- Department of Immunology, Flinders University, Bedford Park, SA, Australia
| | - Jing Jing Wang
- Department of Immunology, SA Pathology (Flinders Medical Centre), Bedford Park, SA, Australia
- Department of Immunology, Flinders University, Bedford Park, SA, Australia
| |
Collapse
|
18
|
Lee AYS, Chataway T, Colella AD, Gordon TP, Wang JJ. Quantitative Mass Spectrometric Analysis of Autoantibodies as a Paradigm Shift in Autoimmune Serology. Front Immunol 2019; 10:2845. [PMID: 31867009 PMCID: PMC6904311 DOI: 10.3389/fimmu.2019.02845] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/19/2019] [Indexed: 12/11/2022] Open
Affiliation(s)
- Adrian Y S Lee
- Department of Immunology, SA Pathology, Flinders Medical Centre, Adelaide, SA, Australia.,College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Tim Chataway
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Alex D Colella
- Department of Immunology, SA Pathology, Flinders Medical Centre, Adelaide, SA, Australia.,College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Tom P Gordon
- Department of Immunology, SA Pathology, Flinders Medical Centre, Adelaide, SA, Australia.,College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Jing J Wang
- Department of Immunology, SA Pathology, Flinders Medical Centre, Adelaide, SA, Australia.,College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
19
|
Adamson PJ, Wang JJ, Anosova NG, Colella AD, Chataway TK, Kleanthous H, Gordon TP, Gordon DL. Proteomic profiling of precipitated Clostridioides difficile toxin A and B antibodies. Vaccine 2019; 38:2077-2087. [PMID: 31718902 DOI: 10.1016/j.vaccine.2019.10.096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/24/2019] [Accepted: 10/29/2019] [Indexed: 01/01/2023]
Abstract
Clostridioides difficile infection is the leading cause of nosocomial diarrhoea globally. Immune responses to toxins produced by C. difficile are important in disease progression and outcome. Here, we analysed the anti-toxin A and anti-toxin B serum antibody proteomes following natural infection or vaccination with a C. difficile toxoid A/toxoid B vaccine using a modified miniaturised proteomic approach based on de novo mass spectrometric sequencing. Analysis of immunoglobulin variable region (IgV) subfamily expression in immunoprecipitated toxin A and toxin B antibodies from four and seven participants of a vaccine trial, respectively, revealed a polyclonal proteome with restricted IGHV, IGKV and IGLV subfamily usage. No dominant IGHV subfamily was observed in the toxin A response, however the dominant anti-toxin B heavy (H)-chain was encoded by IGHV3-23. Light (L)-chain usage was convergent for both anti-toxin A and anti-toxin B proteomes with IGKV3-11, 3-15, 3-20 and 4-1 shared among all subjects in both cohorts. Peptide mapping of common IgV families showed extensive public and private amino acid substitutions. The cohort responses to toxin A and toxin B showed limited similarity in shared IGHV subfamilies. L-chain subfamily usage was more similar in the anti-toxin A and anti-toxin B responses, however the mutational signatures for each subfamily were toxin-dependent. Samples taken both post vaccination (n = 5) or at baseline, indicating previous exposure (n = 2), showed similar anti-toxin B IgV subfamily usage and mutational profiles. In summary, this study provides the first sequence-based proteomic analysis of the antibody response to the major disease-mediating toxins of C. difficile, toxin A and toxin B, and demonstrates that despite the potential for extreme diversity, the immunoglobulin repertoire can raise convergent responses to specific pathogens whether through natural infection or following vaccination.
Collapse
Affiliation(s)
- Penelope J Adamson
- Department of Microbiology and Infectious Diseases, Flinders University and SA Pathology, Flinders Medical Centre, Bedford Park, SA 5042, Australia.
| | - Jing J Wang
- Department of Immunology, Flinders University and SA Pathology, Flinders Medical Centre, Bedford Park, SA 5042, Australia.
| | | | - Alex D Colella
- Flinders Proteomic Facility, Flinders University, Flinders Medical Centre, Bedford Park, SA 5042, Australia.
| | - Timothy K Chataway
- Flinders Proteomic Facility, Flinders University, Flinders Medical Centre, Bedford Park, SA 5042, Australia.
| | | | - Tom P Gordon
- Department of Immunology, Flinders University and SA Pathology, Flinders Medical Centre, Bedford Park, SA 5042, Australia.
| | - David L Gordon
- Department of Microbiology and Infectious Diseases, Flinders University and SA Pathology, Flinders Medical Centre, Bedford Park, SA 5042, Australia.
| |
Collapse
|
20
|
Donlin LT, Park SH, Giannopoulou E, Ivovic A, Park-Min KH, Siegel RM, Ivashkiv LB. Insights into rheumatic diseases from next-generation sequencing. Nat Rev Rheumatol 2019; 15:327-339. [PMID: 31000790 PMCID: PMC6673602 DOI: 10.1038/s41584-019-0217-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Rheumatic diseases have complex aetiologies that are not fully understood, which makes the study of pathogenic mechanisms in these diseases a challenge for researchers. Next-generation sequencing (NGS) and related omics technologies, such as transcriptomics, epigenomics and genomics, provide an unprecedented genome-wide view of gene expression, environmentally responsive epigenetic changes and genetic variation. The integrated application of NGS technologies to samples from carefully phenotyped clinical cohorts of patients has the potential to solve remaining mysteries in the pathogenesis of several rheumatic diseases, to identify new therapeutic targets and to underpin a precision medicine approach to the diagnosis and treatment of rheumatic diseases. This Review provides an overview of the NGS technologies available, showcases important advances in rheumatic disease research already powered by these technologies and highlights NGS approaches that hold particular promise for generating new insights and advancing the field.
Collapse
Affiliation(s)
- Laura T Donlin
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY, USA
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sung-Ho Park
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY, USA
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Eugenia Giannopoulou
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY, USA
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
- Biological Sciences Department, New York City College of Technology, City University of New York, New York, NY, USA
| | - Aleksandra Ivovic
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kyung-Hyun Park-Min
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY, USA
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Richard M Siegel
- Immunoregulation Section, Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lionel B Ivashkiv
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY, USA.
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA.
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
21
|
Wang JJ, Colella AD, Beroukas D, Chataway TK, Gordon TP. Precipitating anti-dsDNA peptide repertoires in lupus. Clin Exp Immunol 2018; 194:273-282. [PMID: 30086185 DOI: 10.1111/cei.13197] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2018] [Indexed: 12/15/2022] Open
Abstract
Anti-double-stranded (ds)DNA autoantibodies are prototypical serological markers of systemic lupus erythematosus (SLE), but little is known about their immunoglobulin variable (IgV) region composition at the level of the secreted (serum) proteome. Here, we use a novel proteomic workflow based on de novo mass spectrometric sequencing of anti-dsDNA precipitins to analyse IgV subfamily expression and mutational signatures of high-affinity, precipitating anti-dsDNA responses. Serum anti-dsDNA proteomes were oligoclonal with shared (public) expression of immunoglobulin (Ig)G heavy chain variable region (IGHV) and kappa chain variable region (IGKV) subfamilies. IgV peptide maps from eight subjects showed extensive public and random (private) amino acid replacement mutations with prominent arginine substitutions across heavy (H)- and light (L)-chains. Shared sets of L-chain complementarity determining region 3 (CDR3) peptides specified by arginine substitutions were sequenced from the dominantly expressed IGKV3-20 subfamily, with changes in expression levels of a clonal L-chain CDR3 peptide by quantitative multiple reaction monitoring (MRM) paralleling the rise and fall of anti-dsDNA levels by Farr radioimmunoassays (RIA). The heavily mutated IgV peptide signatures of precipitating anti-dsDNA autoantibody proteomes reflect the strong selective forces that shape humoral anti-dsDNA responses in germinal centres. Direct sequencing of agarose gel precipitins using microlitre volumes of stored sera streamlines the antibody sequencing workflow and is generalizable to other precipitating serum antibodies.
Collapse
Affiliation(s)
- J J Wang
- Department of Immunology, Flinders Medical Centre and Flinders University, SA Pathology, Bedford Park, SA, Australia
| | - A D Colella
- Department of Immunology, Flinders Medical Centre and Flinders University, SA Pathology, Bedford Park, SA, Australia
| | - D Beroukas
- Department of Immunology, Flinders Medical Centre and Flinders University, SA Pathology, Bedford Park, SA, Australia
| | - T K Chataway
- Flinders Proteomics Facility, Flinders University, Bedford Park, SA, Australia
| | - T P Gordon
- Department of Immunology, Flinders Medical Centre and Flinders University, SA Pathology, Bedford Park, SA, Australia
| |
Collapse
|