1
|
Deguine J, Xavier RJ. B cell tolerance and autoimmunity: Lessons from repertoires. J Exp Med 2024; 221:e20231314. [PMID: 39093312 PMCID: PMC11296956 DOI: 10.1084/jem.20231314] [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: 04/25/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
Adaptive immune cell function is regulated by a highly diverse receptor recombined from variable germline-encoded segments that can recognize an almost unlimited array of epitopes. While this diversity enables the recognition of any pathogen, it also poses a risk of self-recognition, leading to autoimmunity. Many layers of regulation are present during both the generation and activation of B cells to prevent this phenomenon, although they are evidently imperfect. In recent years, our ability to analyze immune repertoires at scale has drastically increased, both through advances in sequencing and single-cell analyses. Here, we review the current knowledge on B cell repertoire analyses, focusing on their implication for autoimmunity. These studies demonstrate that a failure of tolerance occurs at multiple independent checkpoints in different autoimmune contexts, particularly during B cell maturation, plasmablast differentiation, and within germinal centers. These failures are marked by distinct repertoire features that may be used to identify disease- or patient-specific therapeutic approaches.
Collapse
Affiliation(s)
- Jacques Deguine
- Immunology Program, Broad Institute of Massachusetts Institute of Technology and Harvard , Cambridge, MA, USA
| | - Ramnik J Xavier
- Immunology Program, Broad Institute of Massachusetts Institute of Technology and Harvard , Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School , Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
2
|
Raposo B, Klareskog L, Robinson WH, Malmström V, Grönwall C. The peculiar features, diversity and impact of citrulline-reactive autoantibodies. Nat Rev Rheumatol 2024; 20:399-416. [PMID: 38858604 DOI: 10.1038/s41584-024-01124-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2024] [Indexed: 06/12/2024]
Abstract
Since entering the stage 25 years ago as a highly specific serological biomarker for rheumatoid arthritis, anti-citrullinated protein antibodies (ACPAs) have been a topic of extensive research. This hallmark B cell response arises years before disease onset, displays interpatient autoantigen variability, and is associated with poor clinical outcomes. Technological and scientific advances have revealed broad clonal diversity and intriguing features including high levels of somatic hypermutation, variable-domain N-linked glycosylation, hapten-like peptide interactions, and clone-specific multireactivity to citrullinated, carbamylated and acetylated epitopes. ACPAs have been found in different isotypes and subclasses, in both circulation and tissue, and are secreted by both plasmablasts and long-lived plasma cells. Notably, although some disease-promoting features have been reported, results now demonstrate that certain monoclonal ACPAs therapeutically block arthritis and inflammation in mouse models. A wealth of functional studies using patient-derived polyclonal and monoclonal antibodies have provided evidence for pathogenic and protective effects of ACPAs in the context of arthritis. To understand the roles of ACPAs, one needs to consider their immunological properties by incorporating different facets such as rheumatoid arthritis B cell biology, environmental triggers and chronic antigen exposure. The emerging picture points to a complex role of citrulline-reactive autoantibodies, in which the diversity and dynamics of antibody clones could determine clinical progression and manifestations.
Collapse
Affiliation(s)
- Bruno Raposo
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Klareskog
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - William H Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Vivianne Malmström
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Caroline Grönwall
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
3
|
Hu Y, Huang J, Wang S, Sun X, Wang X, Yu H. Deciphering Autoimmune Diseases: Unveiling the Diagnostic, Therapeutic, and Prognostic Potential of Immune Repertoire Sequencing. Inflammation 2024:10.1007/s10753-024-02079-2. [PMID: 38914737 DOI: 10.1007/s10753-024-02079-2] [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] [Received: 04/30/2024] [Revised: 05/31/2024] [Accepted: 06/08/2024] [Indexed: 06/26/2024]
Abstract
Autoimmune diseases (AIDs) are immune system disorders where the body exhibits an immune response to its own antigens, causing damage to its own tissues and organs. The pathogenesis of AIDs is incompletely understood. However, recent advances in immune repertoire sequencing (IR-seq) technology have opened-up a new avenue to study the IR. These studies have revealed the prevalence in IR alterations, potentially inducing AIDs by disrupting immune tolerance and thereby contributing to our comprehension of AIDs. IR-seq harbors significant potential for the clinical diagnosis, personalized treatment, and prognosis of AIDs. This article reviews the application and progress of IR-seq in diseases, such as multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, and type 1 diabetes, to enhance our understanding of the pathogenesis of AIDs and offer valuable references for the diagnosis and treatment of AIDs.
Collapse
Affiliation(s)
- Yuelin Hu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Jialing Huang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Shuqing Wang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Xin Sun
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Xin Wang
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Hongsong Yu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China.
| |
Collapse
|
4
|
Thomas MA, Naik P, Wang H, Giles JT, Girgis AA, Kim SY, Johnson TP, Curran AM, Crawford JD, Jahanbani S, Bingham CO, Robinson WH, Na CH, Darrah E. The monocyte cell surface is a unique site of autoantigen generation in rheumatoid arthritis. Proc Natl Acad Sci U S A 2024; 121:e2304199121. [PMID: 38630712 PMCID: PMC11047081 DOI: 10.1073/pnas.2304199121] [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: 03/15/2023] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
Although anti-citrullinated protein autoantibodies (ACPAs) are a hallmark serological feature of rheumatoid arthritis (RA), the mechanisms and cellular sources behind the generation of the RA citrullinome remain incompletely defined. Peptidylarginine deiminase IV (PAD4), one of the key enzymatic drivers of citrullination in the RA joint, is expressed by granulocytes and monocytes; however, the subcellular localization and contribution of monocyte-derived PAD4 to the generation of citrullinated autoantigens remain underexplored. In this study, we demonstrate that PAD4 displays a widespread cellular distribution in monocytes, including expression on the cell surface. Surface PAD4 was enzymatically active and capable of citrullinating extracellular fibrinogen and endogenous surface proteins in a calcium dose-dependent manner. Fibrinogen citrullinated by monocyte-surface PAD4 could be specifically recognized over native fibrinogen by a panel of eight human monoclonal ACPAs. Several unique PAD4 substrates were identified on the monocyte surface via mass spectrometry, with citrullination of the CD11b and CD18 components of the Mac-1 integrin complex being the most abundant. Citrullinated Mac-1 was found to be a target of ACPAs in 25% of RA patients, and Mac-1 ACPAs were significantly associated with HLA-DRB1 shared epitope alleles, higher C-reactive protein and IL-6 levels, and more erosive joint damage. Our findings implicate the monocyte cell surface as a unique and consequential site of extracellular and cell surface autoantigen generation in RA.
Collapse
Affiliation(s)
- Mekha A. Thomas
- Division of Rheumatology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD21224
| | - Pooja Naik
- Division of Rheumatology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD21224
| | - Hong Wang
- Division of Rheumatology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD21224
| | - Jon T. Giles
- Division of Rheumatology, Columbia University, College of Physicians and Surgeons, New York, NY10032
| | - Alexander A. Girgis
- Division of Rheumatology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD21224
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21224
| | - Seok-Young Kim
- Department of Neurology, Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Tory P. Johnson
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD20892
| | - Ashley M. Curran
- Division of Rheumatology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD21224
| | - Jonathan D. Crawford
- Division of Rheumatology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD21224
| | - Shaghayegh Jahanbani
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA94304
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA94550
| | - Clifton O. Bingham
- Division of Rheumatology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD21224
| | - William H. Robinson
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA94304
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA94550
| | - Chan Hyun Na
- Department of Neurology, Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Erika Darrah
- Division of Rheumatology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD21224
| |
Collapse
|
5
|
Feng F, Yuen R, Wang Y, Hua A, Kepler TB, Wetzler LM. Characterizing adjuvants' effects at murine immunoglobulin repertoire level. iScience 2024; 27:108749. [PMID: 38269092 PMCID: PMC10805652 DOI: 10.1016/j.isci.2023.108749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 09/29/2023] [Accepted: 12/12/2023] [Indexed: 01/26/2024] Open
Abstract
Generating large-scale, high-fidelity sequencing data is challenging and, furthermore, not much has been done to characterize adjuvants' effects at the repertoire level. Thus, we introduced an IgSeq pipeline that standardized library prep protocols and data analysis functions for accurate repertoire profiling. We then studied systemically effects of CpG and Alum on the Ig heavy chain repertoire using the ovalbumin (OVA) murine model. Ig repertoires of different tissues (spleen and bone marrow) and isotypes (IgG and IgM) were examined and compared in IGHV mutation, gene usage, CDR3 length, clonal diversity, and clonal selection. We found Ig repertoires of different compartments exhibited distinguishable profiles at the non-immunized steady state, and distinctions became more pronounced upon adjuvanted immunizations. Notably, Alum and CpG effects exhibited different tissue- and isotype-preferences. The former led to increased diversity of abundant clones in bone marrow, and the latter promoted the selection of IgG clones in both tissues.
Collapse
Affiliation(s)
- Feng Feng
- Department of Microbiology, Boston University, Boston, MA 02118, USA
| | - Rachel Yuen
- Department of Microbiology, Boston University, Boston, MA 02118, USA
| | - Yumei Wang
- Department of Microbiology, Boston University, Boston, MA 02118, USA
| | - Axin Hua
- Department of Microbiology, Boston University, Boston, MA 02118, USA
| | - Thomas B. Kepler
- Department of Microbiology, Boston University, Boston, MA 02118, USA
- Department of Mathematics and Statistics, Boston University, Boston, MA 02118, USA
| | - Lee M. Wetzler
- Department of Microbiology, Boston University, Boston, MA 02118, USA
- Department of Medicine, Boston University School of Medicine, Boston University, Boston, MA 02118, USA
| |
Collapse
|
6
|
Kyriazopoulou E, Giamarellos-Bourboulis EJ, Akinosoglou K. Biomarkers to guide immunomodulatory treatment: where do we stand? Expert Rev Mol Diagn 2023; 23:945-958. [PMID: 37691280 DOI: 10.1080/14737159.2023.2258063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/20/2023] [Accepted: 09/08/2023] [Indexed: 09/12/2023]
Abstract
INTRODUCTION This review summarizes current progress in the development of biomarkers to guide immunotherapy in oncology, rheumatology, and critical illness. AREAS COVERED An extensive literature search was performed about biomarkers classifying patients' immune responses to guide immunotherapy in oncology, rheumatology, and critical illness. Surface markers, such as programmed death-ligand 1 (PD-L1), genetic biomarkers, such as tumor mutation load, and circulating tumor DNA are biomarkers associated with the effectiveness of immunotherapy in oncology. Genomics, metabolomics, and proteomics play a crucial role in selecting the most suitable therapeutic options for rheumatologic patients. Phenotypes and endotypes are a promising approach to detect critically ill patients with hyper- or hypo-inflammation. Sepsis trials using biomarkers such as ferritin, lymphopenia, HLA-DR expression on monocytes and PD-L1 to guide immunotherapy have been already conducted or are currently ongoing. Immunotherapy in COVID-19 pneumonia, guided by C-reactive protein and soluble urokinase plasminogen activator receptor (suPAR) has improved patient outcomes globally. More research is needed into immunotherapy in other critical conditions. EXPERT OPINION Targeted immunotherapy has improved outcomes in oncology and rheumatology, paving the way for precision medicine in the critically ill. Transcriptomics will play a crucial role in detecting the most suitable candidates for immunomodulation.
Collapse
Affiliation(s)
- Evdoxia Kyriazopoulou
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | | |
Collapse
|
7
|
Baglaenko Y, Wagner C, Bhoj VG, Brodin P, Gershwin ME, Graham D, Invernizzi P, Kidd KK, Korsunsky I, Levy M, Mammen AL, Nizet V, Ramirez-Valle F, Stites EC, Williams MS, Wilson M, Rose NR, Ladd V, Sirota M. Making inroads to precision medicine for the treatment of autoimmune diseases: Harnessing genomic studies to better diagnose and treat complex disorders. CAMBRIDGE PRISMS. PRECISION MEDICINE 2023; 1:e25. [PMID: 38550937 PMCID: PMC10953750 DOI: 10.1017/pcm.2023.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 06/13/2024]
Abstract
Precision Medicine is an emerging approach for disease treatment and prevention that takes into account individual variability in genes, environment, and lifestyle. Autoimmune diseases are those in which the body's natural defense system loses discriminating power between its own cells and foreign cells, causing the body to mistakenly attack healthy tissues. These conditions are very heterogeneous in their presentation and therefore difficult to diagnose and treat. Achieving precision medicine in autoimmune diseases has been challenging due to the complex etiologies of these conditions, involving an interplay between genetic, epigenetic, and environmental factors. However, recent technological and computational advances in molecular profiling have helped identify patient subtypes and molecular pathways which can be used to improve diagnostics and therapeutics. This review discusses the current understanding of the disease mechanisms, heterogeneity, and pathogenic autoantigens in autoimmune diseases gained from genomic and transcriptomic studies and highlights how these findings can be applied to better understand disease heterogeneity in the context of disease diagnostics and therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | - Daniel Graham
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), IRCCS Fondazione San Gerardo dei Tintori, Monza, Italy
| | - Kenneth K. Kidd
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Michael Levy
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew L. Mammen
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, USA
| | - Victor Nizet
- School of Medicine, University of California San Diego, San Diego, CA, USA
| | | | - Edward C. Stites
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Michael Wilson
- Weill Institute for Neurosciences, Department of Neurology, UCSF, San Francisco, CA, USA
| | - Noel R. Rose
- Autoimmune Association, Clinton Township, MI, USA
| | | | - Marina Sirota
- Bakar Computational Health Sciences Institute, UCSF, San Francisco, CA, USA
- Department of Pediatrics, UCSF, San Francisco, CA, USA
| |
Collapse
|
8
|
Moura MC, Thompson GE, Nelson DR, Fussner LA, Hummel AM, Jenne DE, Emerling D, Fervenza FC, Kallenberg CGM, Langford CA, McCune WJ, Merkel PA, Monach PA, Seo P, Spiera RF, St. Clair EW, Ytterberg SR, Stone JH, Robinson WH, Specks U. Activation of a Latent Epitope Causing Differential Binding of Antineutrophil Cytoplasmic Antibodies to Proteinase 3. Arthritis Rheumatol 2023; 75:748-759. [PMID: 36515151 PMCID: PMC10191989 DOI: 10.1002/art.42418] [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: 08/18/2022] [Revised: 11/17/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Proteinase 3 (PR3) is the major antigen for antineutrophil cytoplasmic antibodies (ANCAs) in the systemic autoimmune vasculitis, granulomatosis with polyangiitis (GPA). PR3-targeting ANCAs (PR3-ANCAs) recognize different epitopes on PR3. This study was undertaken to study the effect of mutations on PR3 antigenicity. METHODS The recombinant PR3 variants, iPR3 (clinically used to detect PR3-ANCAs) and iHm5 (containing 3 point mutations in epitopes 1 and 5 generated for epitope mapping studies) immunoassays and serum samples from patients enrolled in ANCA-associated vasculitis (AAV) trials were used to screen for differential PR3-ANCA binding. A patient-derived monoclonal ANCA 518 (moANCA518) that selectively binds to iHm5 within the mutation-free epitope 3 and is distant from the point mutations of iHm5 was used as a gauge for remote epitope activation. Selective binding was determined using inhibition experiments. RESULTS Rather than reduced binding of PR3-ANCAs to iHm5, we found substantially increased binding of the majority of PR3-ANCAs to iHm5 compared to iPR3. This differential binding of PR3-ANCA to iHm5 is similar to the selective moANCA518 binding to iHm5. Binding of iPR3 to monoclonal antibody MCPR3-2 also induced recognition by moANCA518. CONCLUSION The preferential binding of PR3-ANCAs from patients, such as the selective binding of moANCA518 to iHm5, is conferred by increased antigenicity of epitope 3 on iHm5. This can also be induced on iPR3 when captured by monoclonal antibody MCPR2. This previously unrecognized characteristic of PR3-ANCA interactions with its target antigen has implications for studying antibody-mediated autoimmune diseases, understanding variable performance characteristics of immunoassays, and design of potential novel treatment approaches.
Collapse
Affiliation(s)
- Marta Casal Moura
- Mayo Clinic and Foundation, Rochester, MN, USA
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | | | | | - Lynn A. Fussner
- Mayo Clinic and Foundation, Rochester, MN, USA
- Ohio State University, Columbus, OH, USA
| | | | - Dieter E. Jenne
- Max-Planck-Institute for Biological Intelligence, 82152 Martinsried, Germany
| | | | | | | | | | | | | | - Paul A. Monach
- VA Boston Healthcare System, Rheumatology, Boston, MA, USA
| | - Philip Seo
- Johns Hopkins University, Baltimore, MD, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Zhang Y, Li Q, Luo L, Duan C, Shen J, Wang Z. Application of germline antibody features to vaccine development, antibody discovery, antibody optimization and disease diagnosis. Biotechnol Adv 2023; 65:108143. [PMID: 37023966 DOI: 10.1016/j.biotechadv.2023.108143] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023]
Abstract
Although the efficacy and commercial success of vaccines and therapeutic antibodies have been tremendous, designing and discovering new drug candidates remains a labor-, time- and cost-intensive endeavor with high risks. The main challenges of vaccine development are inducing a strong immune response in broad populations and providing effective prevention against a group of highly variable pathogens. Meanwhile, antibody discovery faces several great obstacles, especially the blindness in antibody screening and the unpredictability of the developability and druggability of antibody drugs. These challenges are largely due to poorly understanding of germline antibodies and the antibody responses to pathogen invasions. Thanks to the recent developments in high-throughput sequencing and structural biology, we have gained insight into the germline immunoglobulin (Ig) genes and germline antibodies and then the germline antibody features associated with antigens and disease manifestation. In this review, we firstly outline the broad associations between germline antibodies and antigens. Moreover, we comprehensively review the recent applications of antigen-specific germline antibody features, physicochemical properties-associated germline antibody features, and disease manifestation-associated germline antibody features on vaccine development, antibody discovery, antibody optimization, and disease diagnosis. Lastly, we discuss the bottlenecks and perspectives of current and potential applications of germline antibody features in the biotechnology field.
Collapse
Affiliation(s)
- Yingjie Zhang
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Qing Li
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Liang Luo
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Changfei Duan
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China
| | - Zhanhui Wang
- National Key Laboratory of Veterinary Public Health Security, Beijing Key Laboratory of Detection Technology for Animal-Derived Food, College of Veterinary Medicine, China Agricultural University, 100193 Beijing, People's Republic of China.
| |
Collapse
|
10
|
Fahad AS, Madan B, DeKosky BJ. Bioinformatic Analysis of Natively Paired VH:VL Antibody Repertoires for Antibody Discovery. Methods Mol Biol 2023; 2552:447-463. [PMID: 36346608 DOI: 10.1007/978-1-0716-2609-2_25] [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] [Indexed: 06/16/2023]
Abstract
Next-generation DNA sequencing (NGS) of human antibody repertoires has been extensively implemented to discover novel antibody drugs, to analyze B-cell developmental features, and to investigate antibody responses to infectious diseases and vaccination. Because the antibody repertoire encoded by human B cells is highly diverse, NGS analyses of antibody genes have provided a new window into understanding antibody responses for basic immunology, biopharmaceutical drug discovery, and immunotherapy. However, many antibody discovery protocols analyze the heavy and light chains separately due to the short-read nature of most NGS technologies, whereas paired heavy and light chain data are required for complete antibody characterization. Here, we describe a computational workflow to process millions of paired antibody heavy and light chain DNA sequence reads using the Illumina MiSeq 2x300 NGS platform. In this workflow, we describe raw NGS read processing and initial quality filtering, the annotation and assembly of antibody clonotypes relating to paired heavy and light chain antibody lineages, and the generation of complete heavy+light consensus sequences for the downstream cloning and expression of human antibody proteins.
Collapse
Affiliation(s)
- Ahmed S Fahad
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, USA
| | - Bharat Madan
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, USA
| | - Brandon J DeKosky
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, The University of Kansas, Lawrence, KS, USA.
| |
Collapse
|
11
|
Chriswell ME, Lefferts AR, Clay MR, Hsu AR, Seifert J, Feser ML, Rims C, Bloom MS, Bemis EA, Liu S, Maerz MD, Frank DN, Demoruelle MK, Deane KD, James EA, Buckner JH, Robinson WH, Holers VM, Kuhn KA. Clonal IgA and IgG autoantibodies from individuals at risk for rheumatoid arthritis identify an arthritogenic strain of Subdoligranulum. Sci Transl Med 2022; 14. [PMID: 36288282 PMCID: PMC9804515 DOI: 10.1126/scitranslmed.abn5166 10.1126/scitranslmed.abn5166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The mucosal origins hypothesis of rheumatoid arthritis (RA) proposes a central role for mucosal immune responses in the initiation or perpetuation of the systemic autoimmunity that occurs with disease. However, the connection between the mucosa and systemic autoimmunity in RA remains unclear. Using dual immunoglobulin A (IgA) and IgG family plasmablast-derived monoclonal autoantibodies obtained from peripheral blood of individuals at risk for RA, we identified cross-reactivity between RA-relevant autoantigens and bacterial taxa in the closely related families Lachnospiraceae and Ruminococcaceae. After generating bacterial isolates within the Lachnospiraceae/Ruminococcaceae genus Subdoligranulum from the feces of an individual, we confirmed monoclonal antibody binding and CD4+ T cell activation in individuals with RA compared to control individuals. In addition, when Subdoligranulum isolate 7 but not isolate 1 colonized germ-free mice, it stimulated TH17 cell expansion, serum RA-relevant IgG autoantibodies, and joint swelling reminiscent of early RA, with histopathology characterized by antibody deposition and complement activation. Systemic immune responses were likely due to mucosal invasion along with the generation of colon-isolated lymphoid follicles driving increased fecal and serum IgA by isolate 7, because B and CD4+ T cell depletion not only halted intestinal immune responses but also eliminated detectable clinical disease. In aggregate, these findings demonstrate a mechanism of RA pathogenesis through which a specific intestinal strain of bacteria can drive systemic autoantibody generation and joint-centered antibody deposition and immune activation.
Collapse
Affiliation(s)
- Meagan E. Chriswell
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Adam R. Lefferts
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Michael R. Clay
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Alex Ren Hsu
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Jennifer Seifert
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Marie L. Feser
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Cliff Rims
- Benaroya Research Institute, Seattle, WA 98101
| | - Michelle S. Bloom
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Elizabeth A. Bemis
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Sucai Liu
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | | | - Daniel N. Frank
- Division of Infectious Disease, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - M. Kristen Demoruelle
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kevin D. Deane
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | | | | | - William H. Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - V. Michael Holers
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kristine A. Kuhn
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045,Corresponding Author:
| |
Collapse
|
12
|
Chriswell ME, Lefferts AR, Clay MR, Hsu AR, Seifert J, Feser ML, Rims C, Bloom MS, Bemis EA, Liu S, Maerz MD, Frank DN, Demoruelle MK, Deane KD, James EA, Buckner JH, Robinson WH, Holers VM, Kuhn KA. Clonal IgA and IgG autoantibodies from individuals at risk for rheumatoid arthritis identify an arthritogenic strain of Subdoligranulum. Sci Transl Med 2022; 14:eabn5166. [PMID: 36288282 PMCID: PMC9804515 DOI: 10.1126/scitranslmed.abn5166] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The mucosal origins hypothesis of rheumatoid arthritis (RA) proposes a central role for mucosal immune responses in the initiation or perpetuation of the systemic autoimmunity that occurs with disease. However, the connection between the mucosa and systemic autoimmunity in RA remains unclear. Using dual immunoglobulin A (IgA) and IgG family plasmablast-derived monoclonal autoantibodies obtained from peripheral blood of individuals at risk for RA, we identified cross-reactivity between RA-relevant autoantigens and bacterial taxa in the closely related families Lachnospiraceae and Ruminococcaceae. After generating bacterial isolates within the Lachnospiraceae/Ruminococcaceae genus Subdoligranulum from the feces of an individual, we confirmed monoclonal antibody binding and CD4+ T cell activation in individuals with RA compared to control individuals. In addition, when Subdoligranulum isolate 7 but not isolate 1 colonized germ-free mice, it stimulated TH17 cell expansion, serum RA-relevant IgG autoantibodies, and joint swelling reminiscent of early RA, with histopathology characterized by antibody deposition and complement activation. Systemic immune responses were likely due to mucosal invasion along with the generation of colon-isolated lymphoid follicles driving increased fecal and serum IgA by isolate 7, because B and CD4+ T cell depletion not only halted intestinal immune responses but also eliminated detectable clinical disease. In aggregate, these findings demonstrate a mechanism of RA pathogenesis through which a specific intestinal strain of bacteria can drive systemic autoantibody generation and joint-centered antibody deposition and immune activation.
Collapse
Affiliation(s)
- Meagan E. Chriswell
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Adam R. Lefferts
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Michael R. Clay
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Alex Ren Hsu
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Jennifer Seifert
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Marie L. Feser
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Cliff Rims
- Benaroya Research Institute, Seattle, WA 98101
| | - Michelle S. Bloom
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Elizabeth A. Bemis
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Sucai Liu
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | | | - Daniel N. Frank
- Division of Infectious Disease, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - M. Kristen Demoruelle
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kevin D. Deane
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | | | | | - William H. Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - V. Michael Holers
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kristine A. Kuhn
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045,Corresponding Author:
| |
Collapse
|
13
|
Romão VC, Fonseca JE. Disease mechanisms in preclinical rheumatoid arthritis: A narrative review. Front Med (Lausanne) 2022; 9:689711. [PMID: 36059838 PMCID: PMC9437632 DOI: 10.3389/fmed.2022.689711] [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: 04/01/2021] [Accepted: 08/04/2022] [Indexed: 11/20/2022] Open
Abstract
In the last decades, the concept of preclinical rheumatoid arthritis (RA) has become established. In fact, the discovery that disease mechanisms start years before the onset of clinical RA has been one of the major recent insights in the understanding of RA pathogenesis. In accordance with the complex nature of the disease, preclinical events extend over several sequential phases. In a genetically predisposed host, environmental factors will further increase susceptibility for incident RA. In the initial steps of preclinical disease, immune disturbance mechanisms take place outside the joint compartment, namely in mucosal surfaces, such as the lung, gums or gut. Herein, the persistent immunologic response to altered antigens will lead to breach of tolerance and trigger autoimmunity. In a second phase, the immune response matures and is amplified at a systemic level, with epitope spreading and widening of the autoantibody repertoire. Finally, the synovial and bone compartment are targeted by specific autoantibodies against modified antigens, initiating a local inflammatory response that will eventually culminate in clinically evident synovitis. In this review, we discuss the elaborate disease mechanisms in place during preclinical RA, providing a broad perspective in the light of current evidence.
Collapse
Affiliation(s)
- Vasco C. Romão
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon Academic Medical Centre and European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ERN-ReCONNET), Lisbon, Portugal
- Rheumatology Research Unit, Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - João Eurico Fonseca
- Rheumatology Research Unit, Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
14
|
Jabado O, Maldonado MA, Schiff M, Weinblatt ME, Fleischmann R, Robinson WH, He A, Patel V, Greenfield A, Saini J, Galbraith D, Connolly SE. Differential Changes in ACPA Fine Specificity and Gene Expression in a Randomized Trial of Abatacept and Adalimumab in Rheumatoid Arthritis. Rheumatol Ther 2022; 9:391-409. [PMID: 34878629 PMCID: PMC8964842 DOI: 10.1007/s40744-021-00404-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/17/2021] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION The biologics abatacept and adalimumab have different mechanisms of action (MoAs). We analyzed data from patients with rheumatoid arthritis treated in AMPLE (NCT00929864) to explore the pharmacodynamic effects of abatacept or adalimumab on anti-citrullinated protein antibodies (ACPAs) and gene expression. METHODS AMPLE was a phase IIIb, 2-year, randomized, head-to-head trial of abatacept versus adalimumab. Post hoc analyses of baseline anti-cyclic citrullinated peptide-2 (anti-CCP2, an ACPA surrogate) positive (+) status and ACPA fine-specificity profiles over time, as well as transcriptional profiling (peripheral whole blood), were performed. RESULTS Of 646 patients treated (abatacept, n = 318; adalimumab, n = 328), ACPA and gene expression data were available from 508 and 566 patients, respectively. In anti-CCP2+ patients (n = 388), baseline fine specificities for most ACPAs were highly correlated; over 2 years, levels decreased with abatacept but not adalimumab. By year 2, expression of genes associated with T cell co-stimulation and antibody production was lower for abatacept versus adalimumab; expression of genes associated with proinflammatory signaling was lower for adalimumab versus abatacept. Treatment modulated the expression of T- and B-cell gene signatures, with differences in CD8+ T cells, activated T cells, plasma cells, B cells, natural killer cells (all lower with abatacept versus adalimumab), and polymorphonuclear leukocytes (higher with abatacept versus adalimumab). CONCLUSIONS In AMPLE, despite similar clinical outcomes, data showed that pharmacodynamic/genetic changes after 2 years of abatacept or adalimumab were consistent with drug MoAs. Further assessment of the relationship between such changes and clinical outcomes, including prediction of response, is warranted. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT00929864.
Collapse
Affiliation(s)
| | | | | | | | - Roy Fleischmann
- Metroplex Clinical Research Center and University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Aiqing He
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | | - Sean E Connolly
- Bristol Myers Squibb, B4290 3401 Princeton Pike, Lawrenceville, NJ, 08648, USA.
| |
Collapse
|
15
|
Walker LM, Shiakolas AR, Venkat R, Liu ZA, Wall S, Raju N, Pilewski KA, Setliff I, Murji AA, Gillespie R, Makoah NA, Kanekiyo M, Connors M, Morris L, Georgiev IS. High-Throughput B Cell Epitope Determination by Next-Generation Sequencing. Front Immunol 2022; 13:855772. [PMID: 35401559 PMCID: PMC8984479 DOI: 10.3389/fimmu.2022.855772] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 02/14/2022] [Indexed: 01/12/2023] Open
Abstract
Development of novel technologies for the discovery of human monoclonal antibodies has proven invaluable in the fight against infectious diseases. Among the diverse antibody repertoires elicited by infection or vaccination, often only rare antibodies targeting specific epitopes of interest are of potential therapeutic value. Current antibody discovery efforts are capable of identifying B cells specific for a given antigen; however, epitope specificity information is usually only obtained after subsequent monoclonal antibody production and characterization. Here we describe LIBRA-seq with epitope mapping, a next-generation sequencing technology that enables residue-level epitope determination for thousands of single B cells simultaneously. By utilizing an antigen panel of point mutants within the HIV-1 Env glycoprotein, we identified and confirmed antibodies targeting multiple sites of vulnerability on Env, including the CD4-binding site and the V3-glycan site. LIBRA-seq with epitope mapping is an efficient tool for high-throughput identification of antibodies against epitopes of interest on a given antigen target.
Collapse
Affiliation(s)
- Lauren M. Walker
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Andrea R. Shiakolas
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Rohit Venkat
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Zhaojing Ariel Liu
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Steven Wall
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Nagarajan Raju
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Kelsey A. Pilewski
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Ian Setliff
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Amyn A. Murji
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Rebecca Gillespie
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Nigel A. Makoah
- Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Mark Connors
- National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Lynn Morris
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
- Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| | - Ivelin S. Georgiev
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, TN, United States
- Center for Structural Biology, Vanderbilt University, Nashville, TN, United States
- Program in Computational Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
16
|
Antibody cross-reactivity between casein and myelin-associated glycoprotein results in central nervous system demyelination. Proc Natl Acad Sci U S A 2022; 119:e2117034119. [PMID: 35235454 PMCID: PMC8916005 DOI: 10.1073/pnas.2117034119] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Multiple sclerosis (MS) is the most prevalent autoimmune disease of the central nervous system (CNS), leading to irreversible deficits in young adults. Its pathophysiology is believed to be influenced by environmental determinants. As far back as the 1990s, it had been suggested that there is a correlation between the consumption of cow’s milk and the prevalence of MS. Here, we not only demonstrate that a high percentage of MS patients harbor antibodies to bovine casein but also that antibody cross-reactivity between cow’s milk and CNS antigens can exacerbate demyelination. Our data broaden the current understanding of how diet influences the etiology of MS and set the stage for combining personalized diet plans with disease-modifying treatment strategies. Multiple sclerosis (MS) is a neuroinflammatory demyelinating disease of the central nervous system (CNS) with a high socioeconomic relevance. The pathophysiology of MS, which is both complex and incompletely understood, is believed to be influenced by various environmental determinants, including diet. Since the 1990s, a correlation between the consumption of bovine milk products and MS prevalence has been debated. Here, we show that C57BL/6 mice immunized with bovine casein developed severe spinal cord pathology, in particular, demyelination, which was associated with the deposition of immunoglobulin G. Furthermore, we observed binding of serum from casein-immunized mice to mouse oligodendrocytes in CNS tissue sections and in culture where casein-specific antibodies induced complement-dependent pathology. We subsequently identified myelin-associated glycoprotein (MAG) as a cross-reactive antigenic target. The results obtained from the mouse model were complemented by clinical data showing that serum samples from patients with MS contained significantly higher B cell and antibody reactivity to bovine casein than those from patients with other neurologic diseases. This reactivity correlated with the B cell response to a mixture of CNS antigens and could again be attributed to MAG reactivity. While we acknowledge disease heterogeneity among individuals with MS, we believe that consumption of cow’s milk in a subset of patients with MS who have experienced a previous loss of tolerance to bovine casein may aggravate the disease. Our data suggest that patients with antibodies to bovine casein might benefit from restricting dairy products from their diet.
Collapse
|
17
|
Lanz TV, Brewer RC, Ho PP, Moon JS, Jude KM, Fernandez D, Fernandes RA, Gomez AM, Nadj GS, Bartley CM, Schubert RD, Hawes IA, Vazquez SE, Iyer M, Zuchero JB, Teegen B, Dunn JE, Lock CB, Kipp LB, Cotham VC, Ueberheide BM, Aftab BT, Anderson MS, DeRisi JL, Wilson MR, Bashford-Rogers RJ, Platten M, Garcia KC, Steinman L, Robinson WH. Clonally expanded B cells in multiple sclerosis bind EBV EBNA1 and GlialCAM. Nature 2022; 603:321-327. [PMID: 35073561 PMCID: PMC9382663 DOI: 10.1038/s41586-022-04432-7] [Citation(s) in RCA: 390] [Impact Index Per Article: 195.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 01/14/2022] [Indexed: 11/09/2022]
Abstract
Multiple sclerosis (MS) is a heterogenous autoimmune disease in which autoreactive lymphocytes attack the myelin sheath of the central nervous system. B lymphocytes in the cerebrospinal fluid (CSF) of patients with MS contribute to inflammation and secrete oligoclonal immunoglobulins1,2. Epstein-Barr virus (EBV) infection has been epidemiologically linked to MS, but its pathological role remains unclear3. Here we demonstrate high-affinity molecular mimicry between the EBV transcription factor EBV nuclear antigen 1 (EBNA1) and the central nervous system protein glial cell adhesion molecule (GlialCAM) and provide structural and in vivo functional evidence for its relevance. A cross-reactive CSF-derived antibody was initially identified by single-cell sequencing of the paired-chain B cell repertoire of MS blood and CSF, followed by protein microarray-based testing of recombinantly expressed CSF-derived antibodies against MS-associated viruses. Sequence analysis, affinity measurements and the crystal structure of the EBNA1-peptide epitope in complex with the autoreactive Fab fragment enabled tracking of the development of the naive EBNA1-restricted antibody to a mature EBNA1-GlialCAM cross-reactive antibody. Molecular mimicry is facilitated by a post-translational modification of GlialCAM. EBNA1 immunization exacerbates disease in a mouse model of MS, and anti-EBNA1 and anti-GlialCAM antibodies are prevalent in patients with MS. Our results provide a mechanistic link for the association between MS and EBV and could guide the development of new MS therapies.
Collapse
Affiliation(s)
- Tobias V. Lanz
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA 94305, United States, and the Geriatric Research, Education, and Clinical Centers (GRECC), VA Palo Alto Health Care System, 3801 Miranda Ave, Palo Alto, CA 94304, United States,Department of Neurology, Mannheim Center for Translational Neurosciences (MCTN), Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany,Department of Neurology and National Center for Tumor Diseases, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - R. Camille Brewer
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA 94305, United States, and the Geriatric Research, Education, and Clinical Centers (GRECC), VA Palo Alto Health Care System, 3801 Miranda Ave, Palo Alto, CA 94304, United States
| | - Peggy P. Ho
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Beckman Center for Molecular Medicine, 279 Campus Drive, Stanford, CA 94305, United States
| | - Jae-Seung Moon
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA 94305, United States, and the Geriatric Research, Education, and Clinical Centers (GRECC), VA Palo Alto Health Care System, 3801 Miranda Ave, Palo Alto, CA 94304, United States
| | - Kevin M. Jude
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Beckman Center for Molecular Medicine, 279 Campus Drive, Stanford, CA 94305, United States
| | - Daniel Fernandez
- Stanford ChEM-H Institute, Macromolecular Structure Knowledge Center, 290 Jane Stanford Way, Stanford, CA 94305, United States
| | - Ricardo A. Fernandes
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Beckman Center for Molecular Medicine, 279 Campus Drive, Stanford, CA 94305, United States
| | - Alejandro M. Gomez
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA 94305, United States, and the Geriatric Research, Education, and Clinical Centers (GRECC), VA Palo Alto Health Care System, 3801 Miranda Ave, Palo Alto, CA 94304, United States
| | - Gabriel-Stefan Nadj
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA 94305, United States, and the Geriatric Research, Education, and Clinical Centers (GRECC), VA Palo Alto Health Care System, 3801 Miranda Ave, Palo Alto, CA 94304, United States
| | - Christopher M. Bartley
- Hanna H. Gray Fellow, Howard Hughes Medical Institute, 4000 Jones Bridge Rd, Chevy Chase, MD 20815, United States,Weill Institute for Neurosciences, Department of Psychiatry and Behavioral Sciences, University of California San Francisco, 675 Nelson Rising Ln San Francisco, CA 94158, San Francisco, United States
| | - Ryan D. Schubert
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, 675 Nelson Rising Ln San Francisco, CA 94158, San Francisco, United States
| | - Isobel A. Hawes
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, 675 Nelson Rising Ln San Francisco, CA 94158, San Francisco, United States
| | - Sara E. Vazquez
- Department of Biochemistry and Biophysics, University of California San Francisco, 1700 4th Street, San Francisco, CA 94158, United States
| | - Manasi Iyer
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welsh Road, Stanford, CA, United States
| | - J. Bradley Zuchero
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welsh Road, Stanford, CA, United States
| | - Bianca Teegen
- Institute of Experimental Immunology, Euroimmun AG, Seekamp 31, 23560 Lübeck, Germany
| | - Jeffrey E. Dunn
- Division of Neuroimmunology, Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 213 Quarry Road, Stanford, CA, United States
| | - Christopher B. Lock
- Division of Neuroimmunology, Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 213 Quarry Road, Stanford, CA, United States
| | - Lucas B. Kipp
- Division of Neuroimmunology, Department of Neurology and Neurological Sciences, Stanford University School of Medicine, 213 Quarry Road, Stanford, CA, United States
| | - Victoria C. Cotham
- Department of Biochemistry and Molecular Pharmacology, NYU Perlmutter Cancer Center, and NYU Langone Health Proteomics Laboratory, Division of Advanced Research Technologies, NYU School of Medicine, 430 East 29th St, New York, NY, 10016, United States
| | - Beatrix M. Ueberheide
- Department of Biochemistry and Molecular Pharmacology, NYU Perlmutter Cancer Center, and NYU Langone Health Proteomics Laboratory, Division of Advanced Research Technologies, NYU School of Medicine, 430 East 29th St, New York, NY, 10016, United States
| | - Blake T. Aftab
- Preclinical Science and Translational Medicine, Atara Biotherapeutics, 611 Gateway Blvd South San Francisco, CA 94080, United States
| | - Mark S. Anderson
- Department of Medicine, Diabetes Center, University of California San Francisco, 513 Parnassus Ave, San Francisco, CA 94143, United States
| | - Joseph L. DeRisi
- Department of Biochemistry and Biophysics, University of California San Francisco, 1700 4th Street, San Francisco, CA 94158, United States,Chan Zuckerberg Biohub, University of California San Francisco, 499 Illinois Street, San Francisco, CA 94158, United States
| | - Michael R. Wilson
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, 675 Nelson Rising Ln San Francisco, CA 94158, San Francisco, United States
| | - Rachael J.M. Bashford-Rogers
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Dr, Headington, Oxford OX3 7BN, United Kingdom
| | - Michael Platten
- Department of Neurology, Mannheim Center for Translational Neurosciences (MCTN), Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany,Department of Neurology and National Center for Tumor Diseases, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany,DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - K. Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Beckman Center for Molecular Medicine, 279 Campus Drive, Stanford, CA 94305, United States
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Beckman Center for Molecular Medicine, 279 Campus Drive, Stanford, CA 94305, United States
| | - William H. Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA 94305, United States, and the Geriatric Research, Education, and Clinical Centers (GRECC), VA Palo Alto Health Care System, 3801 Miranda Ave, Palo Alto, CA 94304, United States,Corresponding Author: William H. Robinson, Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA 94305, United States,
| |
Collapse
|
18
|
Jarrell JA, Baker MC, Perugino CA, Liu H, Bloom MS, Maehara T, Wong HH, Lanz TV, Adamska JZ, Kongpachith S, Sokolove J, Stone JH, Pillai SS, Robinson WH. Neutralizing anti-IL-1 receptor antagonist autoantibodies induce inflammatory and fibrotic mediators in IgG4-related disease. J Allergy Clin Immunol 2022; 149:358-368. [PMID: 33974929 PMCID: PMC8573062 DOI: 10.1016/j.jaci.2021.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND IgG4-related disease (IgG4-RD) is a fibroinflammatory condition involving loss of B-cell tolerance and production of autoantibodies. However, the relevant targets and role of these aberrant humoral immune responses are not defined. OBJECTIVE Our aim was to identify novel autoantibodies and autoantigen targets that promote pathogenic responses in IgG4-RD. METHODS We sequenced plasmablast antibody repertoires in patients with IgG4-RD. Representative mAbs were expressed and their specificities characterized by using cytokine microarrays. The role of anti-IL-1 receptor antagonist (IL-1RA) autoantibodies was investigated by using in vitro assays. RESULTS We identified strong reactivity against human IL-1RA by using a clonally expanded plasmablast-derived mAb from a patient with IgG4-RD. Plasma from patients with IgG4-RD exhibited elevated levels of reactivity against IL-1RA compared with plasma from the controls and neutralized IL-1RA activity, resulting in inflammatory and fibrotic mediator production in vitro. IL-1RA was detected in lesional tissues from patients with IgG4-RD. Patients with anti-IL-1RA autoantibodies of the IgG4 subclass had greater numbers of organs affected than did those without anti-IL-1RA autoantibodies. Peptide analyses identified IL-1RA epitopes targeted by anti-IL-1RA antibodies at sites near the IL-1RA/IL-1R interface. Serum from patients with systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) also had elevated levels of anti-IL-1RA autoantibodies compared with those of the controls. CONCLUSION A subset of patients with IgG4-RD have anti-IL-1RA autoantibodies, which promote proinflammatory and profibrotic meditator production via IL-1RA neutralization. These findings support a novel immunologic mechanism underlying the pathogenesis of IgG4-RD. Anti-IL-1RA autoantibodies are also present in a subset of patients with SLE and RA, suggesting a potential common pathway in multiple autoimmune diseases.
Collapse
Affiliation(s)
- Justin A Jarrell
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, Calif; Institute for Immunity, Transplant and Infection, Stanford University, Stanford, Calif; VA Palo Alto Health Care System, Palo Alto, Calif
| | - Matthew C Baker
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, Calif
| | - Cory A Perugino
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Hang Liu
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Michelle S Bloom
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, Calif; Institute for Immunity, Transplant and Infection, Stanford University, Stanford, Calif; VA Palo Alto Health Care System, Palo Alto, Calif
| | - Takashi Maehara
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Heidi H Wong
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, Calif; Institute for Immunity, Transplant and Infection, Stanford University, Stanford, Calif; VA Palo Alto Health Care System, Palo Alto, Calif
| | - Tobias V Lanz
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, Calif; Institute for Immunity, Transplant and Infection, Stanford University, Stanford, Calif; VA Palo Alto Health Care System, Palo Alto, Calif; Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Julia Z Adamska
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, Calif; Institute for Immunity, Transplant and Infection, Stanford University, Stanford, Calif; VA Palo Alto Health Care System, Palo Alto, Calif
| | - Sarah Kongpachith
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, Calif; Institute for Immunity, Transplant and Infection, Stanford University, Stanford, Calif; VA Palo Alto Health Care System, Palo Alto, Calif
| | - Jeremy Sokolove
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, Calif; Institute for Immunity, Transplant and Infection, Stanford University, Stanford, Calif; VA Palo Alto Health Care System, Palo Alto, Calif
| | - John H Stone
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Shiv S Pillai
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - William H Robinson
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, Calif; Institute for Immunity, Transplant and Infection, Stanford University, Stanford, Calif; VA Palo Alto Health Care System, Palo Alto, Calif.
| |
Collapse
|
19
|
Wang Z, Huang J, Xie D, He D, Lu A, Liang C. Toward Overcoming Treatment Failure in Rheumatoid Arthritis. Front Immunol 2021; 12:755844. [PMID: 35003068 PMCID: PMC8732378 DOI: 10.3389/fimmu.2021.755844] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/06/2021] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder characterized by inflammation and bone erosion. The exact mechanism of RA is still unknown, but various immune cytokines, signaling pathways and effector cells are involved. Disease-modifying antirheumatic drugs (DMARDs) are commonly used in RA treatment and classified into different categories. Nevertheless, RA treatment is based on a "trial-and-error" approach, and a substantial proportion of patients show failed therapy for each DMARD. Over the past decades, great efforts have been made to overcome treatment failure, including identification of biomarkers, exploration of the reasons for loss of efficacy, development of sequential or combinational DMARDs strategies and approval of new DMARDs. Here, we summarize these efforts, which would provide valuable insights for accurate RA clinical medication. While gratifying, researchers realize that these efforts are still far from enough to recommend specific DMARDs for individual patients. Precision medicine is an emerging medical model that proposes a highly individualized and tailored approach for disease management. In this review, we also discuss the potential of precision medicine for overcoming RA treatment failure, with the introduction of various cutting-edge technologies and big data.
Collapse
Affiliation(s)
- Zhuqian Wang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Jie Huang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Duoli Xie
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Dongyi He
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai, China
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Chao Liang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
20
|
Dahora LC, Verheul MK, Williams KL, Jin C, Stockdale L, Cavet G, Giladi E, Hill J, Kim D, Leung Y, Bobay BG, Spicer LD, Sawant S, Rijpkema S, Dennison SM, Alam SM, Pollard AJ, Tomaras GD. Salmonella Typhi Vi capsule prime-boost vaccination induces convergent and functional antibody responses. Sci Immunol 2021; 6:eabj1181. [PMID: 34714686 PMCID: PMC9960181 DOI: 10.1126/sciimmunol.abj1181] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Vaccine development to prevent Salmonella Typhi infections has accelerated over the past decade, resulting in licensure of new vaccines, which use the Vi polysaccharide (Vi PS) of the bacterium conjugated to an unrelated carrier protein as the active component. Antibodies elicited by these vaccines are important for mediating protection against typhoid fever. However, the characteristics of protective and functional Vi antibodies are unknown. In this study, we investigated the human antibody repertoire, avidity maturation, epitope specificity, and function after immunization with a single dose of Vi-tetanus toxoid conjugate vaccine (Vi-TT) and after a booster with plain Vi PS (Vi-PS). The Vi-TT prime induced an IgG1-dominant response, whereas the Vi-TT prime followed by the Vi-PS boost induced IgG1 and IgG2 antibody production. B cells from recipients who received both prime and boost showed evidence of convergence, with shared V gene usage and CDR3 characteristics. The detected Vi antibodies showed heterogeneous avidity ranging from 10 μM to 500 pM, with no evidence of affinity maturation after the boost. Vi-specific antibodies mediated Fc effector functions, which correlated with antibody dissociation kinetics but not with association kinetics. We identified antibodies induced by prime and boost vaccines that recognized subdominant epitopes, indicated by binding to the de–O-acetylated Vi backbone. These antibodies also mediated Fc-dependent functions, such as complement deposition and monocyte phagocytosis. Defining strategies on how to broaden epitope targeting for S. Typhi Vi and enriching for antibody Fc functions that protect against typhoid fever will advance the design of high-efficacy Vi vaccines for protection across diverse populations.
Collapse
Affiliation(s)
- Lindsay C. Dahora
- Center for Human Systems Immunology, Duke University, Durham, NC, USA.,Department of Immunology, Duke University, Durham, NC, USA.,Corresponding author. (L.C.D.); (G.D.T.)
| | - Marije K. Verheul
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford and NIHR Oxford Biomedical Research Center, Oxford, UK
| | | | - Celina Jin
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford and NIHR Oxford Biomedical Research Center, Oxford, UK
| | - Lisa Stockdale
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford and NIHR Oxford Biomedical Research Center, Oxford, UK
| | | | | | - Jennifer Hill
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford and NIHR Oxford Biomedical Research Center, Oxford, UK
| | | | | | - Benjamin G. Bobay
- Department of Biochemistry, Duke University, Durham, NC, USA.,Department of Radiology, Duke University, Durham, NC, USA.,Duke University NMR Center, Duke University Medical Center, Durham, NC, USA
| | - Leonard D. Spicer
- Department of Biochemistry, Duke University, Durham, NC, USA.,Department of Radiology, Duke University, Durham, NC, USA.,Duke University NMR Center, Duke University Medical Center, Durham, NC, USA
| | - Sheetal Sawant
- Center for Human Systems Immunology, Duke University, Durham, NC, USA.,Department of Surgery, Duke University, Durham, NC, USA
| | - Sjoerd Rijpkema
- Division of Bacteriology, National Institute of Biological Standards and Control, Potters Bar, UK
| | - S. Moses Dennison
- Center for Human Systems Immunology, Duke University, Durham, NC, USA.,Department of Surgery, Duke University, Durham, NC, USA
| | - S. Munir Alam
- Department of Medicine, Duke University, Durham, NC, USA.,Department of Pathology, Duke University, Durham, NC, USA.,Duke Human Vaccine Institute, Duke University, Durham, NC, USA
| | - Andrew J. Pollard
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford and NIHR Oxford Biomedical Research Center, Oxford, UK
| | - Georgia D. Tomaras
- Center for Human Systems Immunology, Duke University, Durham, NC, USA.,Department of Immunology, Duke University, Durham, NC, USA.,Department of Surgery, Duke University, Durham, NC, USA.,Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA.,Corresponding author. (L.C.D.); (G.D.T.)
| |
Collapse
|
21
|
Żabińska M, Kościelska-Kasprzak K, Krajewska J, Bartoszek D, Augustyniak-Bartosik H, Krajewska M. Immune Cells Profiling in ANCA-Associated Vasculitis Patients-Relation to Disease Activity. Cells 2021; 10:1773. [PMID: 34359942 PMCID: PMC8307495 DOI: 10.3390/cells10071773] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/11/2021] [Indexed: 12/05/2022] Open
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitides (AAV) are a group of necrotizing multiorgan autoimmune vasculitides that predominantly affect small blood vessels and are associated with the presence of ANCAs. The aim was to assess regulatory and effector cell populations accompanied by the suPAR biomarker level and link the so-defined immune state to the AAV disease activity. The research involved a multicomponent description of an immune state encompassing a range of B and T cell subsets such as transitional/regulatory B cells (CD19+CD24++CD38++), naïve B cells (CD19+CD24INTCD38INT), Th17 cells, T regulatory cells (CD4+CD25+FoxP3+) and cytotoxic CD4+CD28- cells by flow cytometry. The suPAR plasma level was measured by ELISA. The results indicate that AAV is associated with an increased suPAR plasma level and immune fingerprint characterized by an expansion of Th17 cells and T cells lacking the costimulatory molecule CD28, accompanied by a decrease of regulatory populations (Tregs and transitional B cells) and NK cells. Decreased numbers of regulatory T cells and transitional B cells were shown to be linked to activation of the AAV disease while the increased suPAR plasma level-to AAV-related deterioration of kidney function. The observed immune fingerprint might be a reflection of peripheral tolerance failure responsible for development and progression of ANCA-associated vasculitides.
Collapse
Affiliation(s)
- Marcelina Żabińska
- Department of Nephrology and Transplantation Medicine, Faculty of Medicine, Wroclaw Medical University, 50-367 Wrocław, Poland; (K.K.-K.); (D.B.); (H.A.-B.); (M.K.)
| | - Katarzyna Kościelska-Kasprzak
- Department of Nephrology and Transplantation Medicine, Faculty of Medicine, Wroclaw Medical University, 50-367 Wrocław, Poland; (K.K.-K.); (D.B.); (H.A.-B.); (M.K.)
| | - Joanna Krajewska
- Department of Otolaryngology, Head and Neck Surgery, Faculty of Medicine, Wroclaw Medical University, 50-367 Wrocław, Poland;
| | - Dorota Bartoszek
- Department of Nephrology and Transplantation Medicine, Faculty of Medicine, Wroclaw Medical University, 50-367 Wrocław, Poland; (K.K.-K.); (D.B.); (H.A.-B.); (M.K.)
| | - Hanna Augustyniak-Bartosik
- Department of Nephrology and Transplantation Medicine, Faculty of Medicine, Wroclaw Medical University, 50-367 Wrocław, Poland; (K.K.-K.); (D.B.); (H.A.-B.); (M.K.)
| | - Magdalena Krajewska
- Department of Nephrology and Transplantation Medicine, Faculty of Medicine, Wroclaw Medical University, 50-367 Wrocław, Poland; (K.K.-K.); (D.B.); (H.A.-B.); (M.K.)
| |
Collapse
|
22
|
Shiakolas AR, Johnson N, Kramer KJ, Suryadevara N, Wrapp D, Periasamy S, Pilewski KA, Raju N, Nargi R, Sutton RE, Walker L, Setliff I, Crowe JE, Bukreyev A, Carnahan RH, McLellan JS, Georgiev IS. Efficient discovery of potently neutralizing SARS-CoV-2 antibodies using LIBRA-seq with ligand blocking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.06.02.446813. [PMID: 34100018 PMCID: PMC8183015 DOI: 10.1101/2021.06.02.446813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
SARS-CoV-2 therapeutic antibody discovery efforts have met with notable success but have been associated with a generally inefficient process, requiring the production and characterization of exceptionally large numbers of candidates for the identification of a small set of leads. Here, we show that incorporating antibody-ligand blocking as part of LIBRA-seq, the high-throughput sequencing platform for antibody discovery, results in efficient identification of ultra-potent neutralizing antibodies against SARS-CoV-2. LIBRA-seq with ligand blocking is a general platform for functional antibody discovery targeting the disruption of antigen-ligand interactions.
Collapse
|
23
|
Wu M, Zhao M, Wu H, Lu Q. Immune repertoire: Revealing the "real-time" adaptive immune response in autoimmune diseases. Autoimmunity 2021; 54:61-75. [PMID: 33650440 DOI: 10.1080/08916934.2021.1887149] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The diversity of the immune repertoire (IR) enables the human immune system to distinguish multifarious antigens (Ags) that humans may encounter throughout life. At the same time, bias or abnormalities in the IR also pay a contribution to the pathogenesis of autoimmune diseases. Rapid advancements in high-throughput sequencing (HTS) technology have ushered in a new era of immune studies, revealing novel molecules and pathways that might result in autoimmunity. In the field of IR, HTS can monitor the immune response status and identify disease-specific immune repertoires. In this review, we summarize updated progress on the mechanisms of the IR and current related studies on four autoimmune diseases, particularly focusing on systemic lupus erythematosus (SLE). These autoimmune diseases can exhibit slightly or significantly skewed IRs and provide novel insights that inform our comprehending of disease pathogenesis and provide potential targets for diagnosis and treatment.
Collapse
Affiliation(s)
- Meiyu Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Ming Zhao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Haijing Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, China.,Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| |
Collapse
|
24
|
Forgacs D, Abreu RB, Sautto GA, Kirchenbaum GA, Drabek E, Williamson KS, Kim D, Emerling DE, Ross TM. Convergent antibody evolution and clonotype expansion following influenza virus vaccination. PLoS One 2021; 16:e0247253. [PMID: 33617543 PMCID: PMC7899375 DOI: 10.1371/journal.pone.0247253] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/03/2021] [Indexed: 12/14/2022] Open
Abstract
Recent advances in high-throughput single cell sequencing have opened up new avenues into the investigation of B cell receptor (BCR) repertoires. In this study, PBMCs were collected from 17 human participants vaccinated with the split-inactivated influenza virus vaccine during the 2016-2017 influenza season. A combination of Immune Repertoire Capture (IRCTM) technology and IgG sequencing was performed on ~7,800 plasmablast (PB) cells and preferential IgG heavy-light chain pairings were investigated. In some participants, a single expanded clonotype accounted for ~22% of their PB BCR repertoire. Approximately 60% (10/17) of participants experienced convergent evolution, possessing public PBs that were elicited independently in multiple participants. Binding profiles of one private and three public PBs confirmed they were all subtype-specific, cross-reactive hemagglutinin (HA) head-directed antibodies. Collectively, this high-resolution antibody repertoire analysis demonstrated the impact evolution can have on BCRs in response to influenza virus vaccination, which can guide future universal influenza prophylactic approaches.
Collapse
Affiliation(s)
- David Forgacs
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States of America
| | - Rodrigo B. Abreu
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States of America
| | - Giuseppe A. Sautto
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States of America
| | - Greg A. Kirchenbaum
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States of America
| | - Elliott Drabek
- Atreca, Inc., South San Francisco, CA, United States of America
| | | | - Dongkyoon Kim
- Atreca, Inc., South San Francisco, CA, United States of America
| | | | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States of America
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States of America
- * E-mail:
| |
Collapse
|
25
|
Pholcharee T, Oyen D, Flores-Garcia Y, Gonzalez-Paez G, Han Z, Williams KL, Volkmuth W, Emerling D, Locke E, Richter King C, Zavala F, Wilson IA. Structural and biophysical correlation of anti-NANP antibodies with in vivo protection against P. falciparum. Nat Commun 2021; 12:1063. [PMID: 33594061 PMCID: PMC7887213 DOI: 10.1038/s41467-021-21221-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 01/13/2021] [Indexed: 01/07/2023] Open
Abstract
The most advanced P. falciparum circumsporozoite protein-based malaria vaccine, RTS,S/AS01 (RTS,S), confers partial protection but with antibody titers that wane relatively rapidly, highlighting the need to elicit more potent and durable antibody responses. Here, we elucidate crystal structures, binding affinities and kinetics, and in vivo protection of eight anti-NANP antibodies derived from an RTS,S phase 2a trial and encoded by three different heavy-chain germline genes. The structures reinforce the importance of homotypic Fab-Fab interactions in protective antibodies and the overwhelmingly dominant preference for a germline-encoded aromatic residue for recognition of the NANP motif. In this study, antibody apparent affinity correlates best with protection in an in vivo mouse model, with the more potent antibodies also recognizing epitopes with repeating secondary structural motifs of type I β- and Asn pseudo 310 turns; such insights can be incorporated into design of more effective immunogens and antibodies for passive immunization.
Collapse
Affiliation(s)
- Tossapol Pholcharee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - David Oyen
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
- Pfizer Inc, San Diego, CA, USA
| | - Yevel Flores-Garcia
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Gonzalo Gonzalez-Paez
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Zhen Han
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
- Wondfo USA Co., Ltd, San Diego, CA, USA
| | | | | | | | - Emily Locke
- PATH's Malaria Vaccine Initiative, Washington, DC, USA
| | | | - Fidel Zavala
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA.
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
26
|
Scott JK, Breden F. The adaptive immune receptor repertoire community as a model for FAIR stewardship of big immunology data. CURRENT OPINION IN SYSTEMS BIOLOGY 2020; 24:71-77. [PMID: 33073065 PMCID: PMC7547575 DOI: 10.1016/j.coisb.2020.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Systems biology involves network-oriented, computational approaches to modeling biological systems through analysis of big biological data. To contribute maximally to scientific progress, big biological data should be FAIR: findable, accessible, interoperable, and reusable. Here, we describe high-throughput sequencing data that characterize the vast diversity of B- and T-cell clones comprising the adaptive immune receptor repertoire (AIRR-seq data) and its contribution to our understanding of COVID-19 (coronavirus disease 19). We describe the accomplishments of the AIRR community, a grass-roots network of interdisciplinary laboratory scientists, bioinformaticians, and policy wonks, in creating and publishing standards, software and repositories for AIRR-seq data based on the FAIR principles.
Collapse
Affiliation(s)
- Jamie K Scott
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Felix Breden
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| |
Collapse
|
27
|
Abstract
The vaccine field is pursuing diverse approaches to translate the molecular insights from analyses of effective antibodies and their targeted epitopes into immunogens capable of eliciting protective immune responses. Here we review current antibody-guided strategies including conformation-based, epitope-based, and lineage-based vaccine approaches, which are yielding promising vaccine candidates now being evaluated in clinical trials. We summarize directions being employed by the field, including the use of sequencing technologies to monitor and track developing immune responses for understanding and improving antibody-based immunity. We review opportunities and challenges to transform powerful new discoveries into safe and effective vaccines, which are encapsulated by vaccine efforts against a variety of pathogens including HIV-1, influenza A virus, malaria parasites, respiratory syncytial virus, and SARS-CoV-2. Overall, this review summarizes the extensive progress that has been made to realize antibody-guided structure-based vaccines, the considerable challenges faced, and the opportunities afforded by recently developed molecular approaches to vaccine development.
Collapse
|
28
|
Antigenic Variation of the Dengue Virus 2 Genotypes Impacts the Neutralization Activity of Human Antibodies in Vaccinees. Cell Rep 2020; 33:108226. [PMID: 33027653 PMCID: PMC7583086 DOI: 10.1016/j.celrep.2020.108226] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 08/04/2020] [Accepted: 09/11/2020] [Indexed: 11/30/2022] Open
Abstract
Dengue virus (DENV) infects an estimated 390 million people each year worldwide. As tetravalent DENV vaccines have variable efficacy against DENV serotype 2 (DENV2), we evaluated the role of genetic diversity within the pre-membrane (prM) and envelope (E) proteins of DENV2 on vaccine performance. We generated a recombinant DENV2 genotype variant panel with contemporary prM and E isolates that are representative of global genetic diversity. The DENV2 genotype variants differ in growth kinetics, morphology, and virion stability. Importantly, the DENV2 genotypic variants are differentially neutralized by monoclonal antibodies, polyclonal serum neutralizing antibodies from DENV2-infected human subjects, and vaccine-elicited antibody responses from the TV003 NIH DENV2 monovalent and DENV tetravalent vaccines. We conclude that DENV2 prM and E genetic diversity significantly modulates antibody neutralization activity. These findings have important implications for dengue vaccines, which are being developed under the assumption that intraserotype variation has minimal impact on neutralizing antibodies. Martinez et al. demonstrate that dengue virus serotype 2 (DENV2) genetic variation modulates neutralizing antibody activity from infection and vaccination. This observation underlines that genotypic variation impacts dengue virus 2 evasion from humoral immunity, suggesting that intraserotype genotypic variation should be considered in designing dengue vaccines.
Collapse
|
29
|
Tanno H, McDaniel JR, Stevens CA, Voss WN, Li J, Durrett R, Lee J, Gollihar J, Tanno Y, Delidakis G, Pothukuchy A, Ellefson JW, Goronzy JJ, Maynard JA, Ellington AD, Ippolito GC, Georgiou G. A facile technology for the high-throughput sequencing of the paired VH:VL and TCRβ:TCRα repertoires. SCIENCE ADVANCES 2020; 6:eaay9093. [PMID: 32426460 PMCID: PMC7176429 DOI: 10.1126/sciadv.aay9093] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 01/27/2020] [Indexed: 05/27/2023]
Abstract
Natively paired sequencing (NPS) of B cell receptors [variable heavy (VH) and light (VL)] and T cell receptors (TCRb and TCRa) is essential for the understanding of adaptive immunity in health and disease. Despite many recent technical advances, determining the VH:VL or TCRb:a repertoire with high accuracy and throughput remains challenging. We discovered that the recently engineered xenopolymerase, RTX, is exceptionally resistant to cell lysate inhibition in single-cell emulsion droplets. We capitalized on the characteristics of this enzyme to develop a simple, rapid, and inexpensive in-droplet overlap extension reverse transcription polymerase chain reaction method for NPS not requiring microfluidics or other specialized equipment. Using this technique, we obtained high yields (5000 to >20,000 per sample) of paired VH:VL or TCRb:a clonotypes at low cost. As a demonstration, we performed NPS on peripheral blood plasmablasts and T follicular helper cells following seasonal influenza vaccination and discovered high-affinity influenza-specific antibodies and TCRb:a.
Collapse
Affiliation(s)
- Hidetaka Tanno
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA
| | - Jonathan R. McDaniel
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA
| | | | - William N. Voss
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA
| | - Jie Li
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA
| | - Russell Durrett
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Jiwon Lee
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Jimmy Gollihar
- Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX, USA
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
- U.S. Army Research Laboratory South, Austin, TX, USA
| | - Yuri Tanno
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA
| | - George Delidakis
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Arti Pothukuchy
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Jared W. Ellefson
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA
- Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX, USA
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Jörg J. Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Jennifer A. Maynard
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Andrew D. Ellington
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA
- Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX, USA
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Gregory C. Ippolito
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - George Georgiou
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, USA
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
30
|
Oyen D, Torres JL, Aoto PC, Flores-Garcia Y, Binter Š, Pholcharee T, Carroll S, Reponen S, Wash R, Liang Q, Lemiale F, Locke E, Bradley A, King CR, Emerling D, Kellam P, Zavala F, Ward AB, Wilson IA. Structure and mechanism of monoclonal antibody binding to the junctional epitope of Plasmodium falciparum circumsporozoite protein. PLoS Pathog 2020; 16:e1008373. [PMID: 32150583 PMCID: PMC7082059 DOI: 10.1371/journal.ppat.1008373] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 03/19/2020] [Accepted: 01/31/2020] [Indexed: 11/21/2022] Open
Abstract
Lasting protection has long been a goal for malaria vaccines. The major surface antigen on Plasmodium falciparum sporozoites, the circumsporozoite protein (PfCSP), has been an attractive target for vaccine development and most protective antibodies studied to date interact with the central NANP repeat region of PfCSP. However, it remains unclear what structural and functional characteristics correlate with better protection by one antibody over another. Binding to the junctional region between the N-terminal domain and central NANP repeats has been proposed to result in superior protection: this region initiates with the only NPDP sequence followed immediately by NANP. Here, we isolated antibodies in Kymab mice immunized with full-length recombinant PfCSP and two protective antibodies were selected for further study with reactivity against the junctional region. X-ray and EM structures of two monoclonal antibodies, mAb667 and mAb668, shed light on their differential affinity and specificity for the junctional region. Importantly, these antibodies also bind to the NANP repeat region with equal or better affinity. A comparison with an NANP-only binding antibody (mAb317) revealed roughly similar but statistically distinct levels of protection against sporozoite challenge in mouse liver burden models, suggesting that junctional antibody protection might relate to the ability to also cross-react with the NANP repeat region. Our findings indicate that additional efforts are necessary to isolate a true junctional antibody with no or much reduced affinity to the NANP region to elucidate the role of the junctional epitope in protection. The circumsporozoite protein (CSP) of Plasmodium falciparum malaria has been the foundation for the design of transmission blocking malaria vaccines. To date, the most promising CSP-based vaccine candidate is RTS,S, which consists of the central repeating NANP amino-acid sequence and the C-terminal domain of CSP fused to hepatitis B surface antigen that assembles into virus-like particles. Potential shortcomings of RTS,S includes the lack of other potential CSP epitopes such as the junctional epitope, which is located between the N-terminal domain of CSP and the start of the NANP repeat region. Here, we elicited antibodies against full-length CSP and screened for junctional epitope binding. We then used an array of biophysical techniques to elucidate the nature of the binding and tested the level of two protective antibodies in a mouse challenge model. Although the antibodies were able to bind both junctional and NANP epitopes, the in vivo data showed distinct levels of protection between themselves and also to an NANP-only binder. Our data suggest that their protection ability may be related to the strong cross-reactivity with NANP epitopes. Since all reported junctional antibodies to date have dual-specificity, we suggest that a true junctional binder with no or very low NANP affinity, if one can be found, is essential to evaluate the contribution of the junctional epitope to protection.
Collapse
Affiliation(s)
- David Oyen
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Jonathan L. Torres
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Phillip C. Aoto
- Department of Pharmacology, University of California at San Diego, La Jolla, California, United States of America
| | - Yevel Flores-Garcia
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Špela Binter
- Kymab Ltd., The Bennet Building (B930), Babraham Research Campus, Cambridge, United Kingdom
| | - Tossapol Pholcharee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Sean Carroll
- Atreca Inc., South San Francisco, California, United States of America
| | - Sini Reponen
- Atreca Inc., South San Francisco, California, United States of America
| | - Rachael Wash
- Kymab Ltd., The Bennet Building (B930), Babraham Research Campus, Cambridge, United Kingdom
| | - Qi Liang
- Kymab Ltd., The Bennet Building (B930), Babraham Research Campus, Cambridge, United Kingdom
| | - Franck Lemiale
- PATH’s Malaria Vaccine Initiative, PATH Center for Vaccine Innovation and Access, Washington, United States of America
| | - Emily Locke
- PATH’s Malaria Vaccine Initiative, PATH Center for Vaccine Innovation and Access, Washington, United States of America
| | - Allan Bradley
- Kymab Ltd., The Bennet Building (B930), Babraham Research Campus, Cambridge, United Kingdom
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - C. Richter King
- PATH’s Malaria Vaccine Initiative, PATH Center for Vaccine Innovation and Access, Washington, United States of America
| | - Daniel Emerling
- Atreca Inc., South San Francisco, California, United States of America
| | - Paul Kellam
- Kymab Ltd., The Bennet Building (B930), Babraham Research Campus, Cambridge, United Kingdom
- Department of Infectious Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Fidel Zavala
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Andrew B. Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, United States of America
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
31
|
Abstract
Cells, the basic units of life, have striking differences at transcriptomic, proteomic and epigenomic levels across tissues, organs, organ systems and organisms. The coordination of individual immune cells is essential for the generation of effective immune responses to pathogens while immune tolerance is maintained to protect the host. In rheumatic diseases, when immune responses are dysregulated, pathologically important cells might represent only a small fraction of the immune system. Interrogation of the contributions of individual immune cells to pathogenesis and disease progression should therefore reveal important insights into the complicated aetiology of rheumatic diseases. Technological advances are enabling the high-dimensional dissection of single cells at multiple omics levels, which could facilitate the identification of dysregulated molecular mechanisms in patients with rheumatic diseases and the discovery of new therapeutic targets and biomarkers. The single-cell technologies that have been developed over the past decade and the experimental platforms that enable multi-omics integrative analyses have already made inroads into immunology-related fields of study and have potential for use in rheumatology. Layers of omics data derived from single cells are likely to fundamentally change our understanding of the molecular pathways that underpin the pathogenesis of rheumatic diseases.
Collapse
|
32
|
Elliott SE, Kongpachith S, Lingampalli N, Adamska JZ, Cannon BJ, Blum LK, Bloom MS, Henkel M, McGeachy MJ, Moreland LW, Robinson WH. B cells in rheumatoid arthritis synovial tissues encode focused antibody repertoires that include antibodies that stimulate macrophage TNF-α production. Clin Immunol 2020; 212:108360. [PMID: 32035179 DOI: 10.1016/j.clim.2020.108360] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/04/2020] [Accepted: 02/04/2020] [Indexed: 01/13/2023]
Abstract
Rheumatoid arthritis (RA) is characterized by the production of anti-citrullinated protein antibodies (ACPAs). To gain insights into the relationship between ACPA-expressing B cells in peripheral blood (PB) and synovial tissue (ST), we sequenced the B cell repertoire in paired PB and ST samples from five individuals with established, ACPA+ RA. Bioinformatics analysis of paired heavy- and light-chain sequences revealed clonally-related family members shared between PB and ST. ST-derived antibody repertoires exhibited reduced diversity and increased normalized clonal family size compared to PB-derived repertoires. Functional characterization showed that seven recombinant antibodies (rAbs) expressed from subject-derived sequences from both compartments bound citrullinated antigens and immune complexes (ICs) formed using one ST-derived rAb stimulated macrophage TNF-α production. Our findings demonstrate B cell trafficking between PB and ST in subjects with RA and ST repertoires include B cells that encode ACPA capable of forming ICs that stimulate cellular responses implicated in RA pathogenesis.
Collapse
Affiliation(s)
- Serra E Elliott
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, United States of America; VA Palo Alto Health Care System, Palo Alto, CA, United States of America
| | - Sarah Kongpachith
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, United States of America; VA Palo Alto Health Care System, Palo Alto, CA, United States of America
| | - Nithya Lingampalli
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, United States of America; VA Palo Alto Health Care System, Palo Alto, CA, United States of America
| | - Julia Z Adamska
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, United States of America; VA Palo Alto Health Care System, Palo Alto, CA, United States of America
| | - Bryan J Cannon
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, United States of America; VA Palo Alto Health Care System, Palo Alto, CA, United States of America
| | - Lisa K Blum
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, United States of America; VA Palo Alto Health Care System, Palo Alto, CA, United States of America
| | - Michelle S Bloom
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, United States of America; VA Palo Alto Health Care System, Palo Alto, CA, United States of America
| | - Matthew Henkel
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Mandy J McGeachy
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Larry W Moreland
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - William H Robinson
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, United States of America; VA Palo Alto Health Care System, Palo Alto, CA, United States of America.
| |
Collapse
|
33
|
Setliff I, Shiakolas AR, Pilewski KA, Murji AA, Mapengo RE, Janowska K, Richardson S, Oosthuysen C, Raju N, Ronsard L, Kanekiyo M, Qin JS, Kramer KJ, Greenplate AR, McDonnell WJ, Graham BS, Connors M, Lingwood D, Acharya P, Morris L, Georgiev IS. High-Throughput Mapping of B Cell Receptor Sequences to Antigen Specificity. Cell 2019; 179:1636-1646.e15. [PMID: 31787378 DOI: 10.1016/j.cell.2019.11.003] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/28/2019] [Accepted: 10/31/2019] [Indexed: 12/15/2022]
Abstract
B cell receptor (BCR) sequencing is a powerful tool for interrogating immune responses to infection and vaccination, but it provides limited information about the antigen specificity of the sequenced BCRs. Here, we present LIBRA-seq (linking B cell receptor to antigen specificity through sequencing), a technology for high-throughput mapping of paired heavy- and light-chain BCR sequences to their cognate antigen specificities. B cells are mixed with a panel of DNA-barcoded antigens so that both the antigen barcode(s) and BCR sequence are recovered via single-cell next-generation sequencing. Using LIBRA-seq, we mapped the antigen specificity of thousands of B cells from two HIV-infected subjects. The predicted specificities were confirmed for a number of HIV- and influenza-specific antibodies, including known and novel broadly neutralizing antibodies. LIBRA-seq will be an integral tool for antibody discovery and vaccine development efforts against a wide range of antigen targets.
Collapse
Affiliation(s)
- Ian Setliff
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Program in Chemical and Physical Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Andrea R Shiakolas
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kelsey A Pilewski
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Amyn A Murji
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rutendo E Mapengo
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa
| | - Katarzyna Janowska
- Division of Structural Biology, Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Simone Richardson
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa; Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Charissa Oosthuysen
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa; Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Nagarajan Raju
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Larance Ronsard
- Ragon Institute of Massachusetts General Hospital, Harvard and Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Juliana S Qin
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kevin J Kramer
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Allison R Greenplate
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Wyatt J McDonnell
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Translational and Clinical Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Mark Connors
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Daniel Lingwood
- Ragon Institute of Massachusetts General Hospital, Harvard and Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Priyamvada Acharya
- Division of Structural Biology, Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC 27710, USA; Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Lynn Morris
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa; Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa; Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban 4041, South Africa
| | - Ivelin S Georgiev
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, TN 37232, USA; Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
34
|
Steen J, Forsström B, Sahlström P, Odowd V, Israelsson L, Krishnamurthy A, Badreh S, Mathsson Alm L, Compson J, Ramsköld D, Ndlovu W, Rapecki S, Hansson M, Titcombe PJ, Bang H, Mueller DL, Catrina AI, Grönwall C, Skriner K, Nilsson P, Lightwood D, Klareskog L, Malmström V. Recognition of Amino Acid Motifs, Rather Than Specific Proteins, by Human Plasma Cell-Derived Monoclonal Antibodies to Posttranslationally Modified Proteins in Rheumatoid Arthritis. Arthritis Rheumatol 2019; 71:196-209. [PMID: 30152202 PMCID: PMC6563427 DOI: 10.1002/art.40699] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 08/23/2018] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Antibodies against posttranslationally modified proteins are a hallmark of rheumatoid arthritis (RA), but the emergence and pathogenicity of these autoantibodies are still incompletely understood. The aim of this study was to analyze the antigen specificities and mutation patterns of monoclonal antibodies (mAb) derived from RA synovial plasma cells and address the question of antigen cross-reactivity. METHODS IgG-secreting cells were isolated from RA synovial fluid, and the variable regions of the immunoglobulins were sequenced (n = 182) and expressed in full-length mAb (n = 93) and also as germline-reverted versions. The patterns of reactivity with 53,019 citrullinated peptides and 49,211 carbamylated peptides and the potential of the mAb to promote osteoclastogenesis were investigated. RESULTS Four unrelated anti-citrullinated protein autoantibodies (ACPAs), of which one was clonally expanded, were identified and found to be highly somatically mutated in the synovial fluid of a patient with RA. The ACPAs recognized >3,000 unique peptides modified by either citrullination or carbamylation. This highly multireactive autoantibody feature was replicated for Ig sequences derived from B cells from the peripheral blood of other RA patients. The plasma cell-derived mAb were found to target distinct amino acid motifs and partially overlapping protein targets. They also conveyed different effector functions as revealed in an osteoclast activation assay. CONCLUSION These findings suggest that the high level of cross-reactivity among RA autoreactive B cells is the result of different antigen encounters, possibly at different sites and at different time points. This is consistent with the notion that RA is initiated in one context, such as in the mucosal organs, and thereafter targets other sites, such as the joints.
Collapse
Affiliation(s)
- Johanna Steen
- Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | - Peter Sahlström
- Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden, and Charité Univeristätsmedizin, Berlin, Germany
| | | | - Lena Israelsson
- Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | - Sara Badreh
- Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | | | - Daniel Ramsköld
- Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | | | - Monika Hansson
- Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Philip J Titcombe
- Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden, and University of Minnesota Medical School, Minneapolis
| | | | | | - Anca I Catrina
- Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Caroline Grönwall
- Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | - Peter Nilsson
- KTH Royal Institute of Technology, Stockholm, Sweden
| | | | - Lars Klareskog
- Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Vivianne Malmström
- Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
35
|
Cashman KS, Jenks SA, Woodruff MC, Tomar D, Tipton CM, Scharer CD, Lee EH, Boss JM, Sanz I. Understanding and measuring human B-cell tolerance and its breakdown in autoimmune disease. Immunol Rev 2019; 292:76-89. [PMID: 31755562 PMCID: PMC6935423 DOI: 10.1111/imr.12820] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022]
Abstract
The maintenance of immunological tolerance of B lymphocytes is a complex and critical process that must be implemented as to avoid the detrimental development of autoreactivity and possible autoimmunity. Murine models have been invaluable to elucidate many of the key components in B-cell tolerance; however, translation to human homeostatic and pathogenic immune states can be difficult to assess. Functional autoreactive, flow cytometric, and single-cell cloning assays have proven to be critical in deciphering breaks in B-cell tolerance within autoimmunity; however, newer approaches to assess human B-cell tolerance may prove to be vital in the further exploration of underlying tolerance defects. In this review, we supply a comprehensive overview of human immune tolerance checkpoints with associated mechanisms of enforcement, and highlight current and future methodologies which are likely to benefit future studies into the mechanisms that become defective in human autoimmune conditions.
Collapse
Affiliation(s)
- Kevin S. Cashman
- Department of Medicine, Division of Rheumatology, Emory University, Atlanta, Georgia, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
| | - Scott A. Jenks
- Department of Medicine, Division of Rheumatology, Emory University, Atlanta, Georgia, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
| | - Matthew C. Woodruff
- Department of Medicine, Division of Rheumatology, Emory University, Atlanta, Georgia, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
| | - Deepak Tomar
- Department of Medicine, Division of Rheumatology, Emory University, Atlanta, Georgia, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
| | - Christopher M. Tipton
- Department of Medicine, Division of Rheumatology, Emory University, Atlanta, Georgia, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
| | - Christopher D. Scharer
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Eun-Hyung Lee
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Emory University, Atlanta, Georgia, USA
| | - Jeremy M. Boss
- Department of Microbiology and Immunology, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Ignacio Sanz
- Department of Medicine, Division of Rheumatology, Emory University, Atlanta, Georgia, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
36
|
Pang YP, Casal Moura M, Thompson GE, Nelson DR, Hummel AM, Jenne DE, Emerling D, Volkmuth W, Robinson WH, Specks U. Remote Activation of a Latent Epitope in an Autoantigen Decoded With Simulated B-Factors. Front Immunol 2019; 10:2467. [PMID: 31708920 PMCID: PMC6823208 DOI: 10.3389/fimmu.2019.02467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/03/2019] [Indexed: 11/13/2022] Open
Abstract
Mutants of a catalytically inactive variant of Proteinase 3 (PR3)—iPR3-Val103 possessing a Ser195Ala mutation relative to wild-type PR3-Val103—offer insights into how autoantigen PR3 interacts with antineutrophil cytoplasmic antibodies (ANCAs) in granulomatosis with polyangiitis (GPA) and whether such interactions can be interrupted. Here we report that iHm5-Val103, a triple mutant of iPR3-Val103, bound a monoclonal antibody (moANCA518) from a GPA patient on an epitope remote from the mutation sites, whereas the corresponding epitope of iPR3-Val103 was latent to moANCA518. Simulated B-factor analysis revealed that the binding of moANCA518 to iHm5-Val103 was due to increased main-chain flexibility of the latent epitope caused by remote mutations, suggesting rigidification of epitopes with therapeutics to alter pathogenic PR3·ANCA interactions as new GPA treatments.
Collapse
Affiliation(s)
- Yuan-Ping Pang
- Computer-Aided Molecular Design Laboratory, Mayo Clinic, Rochester, MN, United States
| | - Marta Casal Moura
- Thoracic Disease Research Unit, Mayo Clinic, Rochester, MN, United States
| | - Gwen E Thompson
- Thoracic Disease Research Unit, Mayo Clinic, Rochester, MN, United States
| | - Darlene R Nelson
- Thoracic Disease Research Unit, Mayo Clinic, Rochester, MN, United States
| | - Amber M Hummel
- Thoracic Disease Research Unit, Mayo Clinic, Rochester, MN, United States
| | - Dieter E Jenne
- Comprehensive Pneumology Center, Helmholtz Zentrum München & Max-Planck Institute for Neuroimmunology, Martinsried, Germany
| | | | | | - William H Robinson
- Department of Medicine, Stanford University, Palo Alto, CA, United States
| | - Ulrich Specks
- Thoracic Disease Research Unit, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
37
|
McLeod B, Miura K, Scally SW, Bosch A, Nguyen N, Shin H, Kim D, Volkmuth W, Rämisch S, Chichester JA, Streatfield S, Woods C, Schief WR, Emerling D, King CR, Julien JP. Potent antibody lineage against malaria transmission elicited by human vaccination with Pfs25. Nat Commun 2019; 10:4328. [PMID: 31551421 PMCID: PMC6760140 DOI: 10.1038/s41467-019-11980-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 08/14/2019] [Indexed: 01/13/2023] Open
Abstract
Transmission-blocking vaccines have the potential to be key contributors to malaria elimination. Such vaccines elicit antibodies that inhibit parasites during their development in Anopheles mosquitoes, thus breaking the cycle of transmission. To date, characterization of humoral responses to Plasmodium falciparum transmission-blocking vaccine candidate Pfs25 has largely been conducted in pre-clinical models. Here, we present molecular analyses of human antibody responses generated in a clinical trial evaluating Pfs25 vaccination. From a collection of monoclonal antibodies with transmission-blocking activity, we identify the most potent transmission-blocking antibody yet described against Pfs25; 2544. The interactions of 2544 and three other antibodies with Pfs25 are analyzed by crystallography to understand structural requirements for elicitation of human transmission-blocking responses. Our analyses provide insights into Pfs25 immunogenicity and epitope potency, and detail an affinity maturation pathway for a potent transmission-blocking antibody in humans. Our findings can be employed to guide the design of improved malaria transmission-blocking vaccines. Pfs25 is a transmission-blocking vaccine candidate for Plasmodium. Here, McLeod et al. analyze the antibody response to Pfs25 in sera from a clinical trial evaluating a Pfs25 vaccine candidate, identify a potent transmission-blocking antibody and determine recognized epitopes on Pfs25.
Collapse
Affiliation(s)
- Brandon McLeod
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON, M5G 0A4, Canada.,Department of Biochemistry, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD, 20852, USA
| | - Stephen W Scally
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Alexandre Bosch
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Ngan Nguyen
- Atreca, 500 Saginaw Drive, Redwood City, CA, 94063-4750, USA
| | - Hanjun Shin
- Atreca, 500 Saginaw Drive, Redwood City, CA, 94063-4750, USA
| | - Dongkyoon Kim
- Atreca, 500 Saginaw Drive, Redwood City, CA, 94063-4750, USA
| | - Wayne Volkmuth
- Atreca, 500 Saginaw Drive, Redwood City, CA, 94063-4750, USA
| | - Sebastian Rämisch
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jessica A Chichester
- Gene Therapy Program & Orphan Disease Center, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Stephen Streatfield
- Fraunhofer USA Center for Molecular Biotechnology CMB, 9 Innovation Way, Newark, DE, 19711, USA
| | - Colleen Woods
- PATH's Malaria Vaccine Initiative, 455 Massachusetts Avenue NW Suite 1000, Washington, DC, 20001, USA
| | - William R Schief
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Daniel Emerling
- Atreca, 500 Saginaw Drive, Redwood City, CA, 94063-4750, USA
| | - C Richter King
- PATH's Malaria Vaccine Initiative, 455 Massachusetts Avenue NW Suite 1000, Washington, DC, 20001, USA
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, 686 Bay Street, Toronto, ON, M5G 0A4, Canada. .,Department of Biochemistry, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada. .,Department of Immunology, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
38
|
Lanz TV, Pröbstel AK, Mildenberger I, Platten M, Schirmer L. Single-Cell High-Throughput Technologies in Cerebrospinal Fluid Research and Diagnostics. Front Immunol 2019; 10:1302. [PMID: 31244848 PMCID: PMC6579921 DOI: 10.3389/fimmu.2019.01302] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 05/22/2019] [Indexed: 01/08/2023] Open
Abstract
High-throughput single-cell technologies have recently emerged as essential tools in biomedical research with great potential for clinical pathology when studying liquid and solid biopsies. We provide an update on current single-cell methods in cerebrospinal fluid research and diagnostics, focusing on high-throughput cell-type specific proteomic and genomic technologies. Proteomic methods comprising flow cytometry and mass cytometry as well as genomic approaches including immune cell repertoire and single-cell transcriptomic studies are critically reviewed and future directions discussed.
Collapse
Affiliation(s)
- Tobias V. Lanz
- Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Anne-Katrin Pröbstel
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
- Departments of Medicine and Biomedicine, Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Iris Mildenberger
- Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lucas Schirmer
- Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
39
|
Nivarthi UK, Tu HA, Delacruz MJ, Swanstrom J, Patel B, Durbin AP, Whitehead SS, Pierce KK, Kirkpatrick BD, Baric RS, Nguyen N, Emerling DE, de Silva AM, Diehl SA. Longitudinal analysis of acute and convalescent B cell responses in a human primary dengue serotype 2 infection model. EBioMedicine 2019; 41:465-478. [PMID: 30857944 PMCID: PMC6444124 DOI: 10.1016/j.ebiom.2019.02.060] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/18/2019] [Accepted: 02/28/2019] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Acute viral infections induce a rapid and transient increase in antibody-secreting plasmablasts. At convalescence, memory B cells (MBC) and long-lived plasma cells (LLPC) are responsible for long-term humoral immunity. Following an acute viral infection, the specific properties and relationships between antibodies produced by these B cell compartments are poorly understood. METHODS We utilized a controlled human challenge model of primary dengue virus serotype 2 (DENV2) infection to study acute and convalescent B-cell responses. FINDINGS The level of DENV2 replication was correlated with the magnitude of the plasmablast response. Functional analysis of plasmablast-derived monoclonal antibodies showed that the DENV2-specific response was dominated by cells producing DENV2 serotype-specific antibodies. DENV2-neutralizing antibodies targeted quaternary structure epitopes centered on domain III of the viral envelope protein (EDIII). Functional analysis of MBC and serum antibodies from the same subjects six months post-challenge revealed maintenance of the serotype-specific response in both compartments. The serum response mainly targeted DENV2 serotype-specific epitopes on EDIII. INTERPRETATION Our data suggest overall functional alignment of DENV2-specific responses from the plasmablast, through the MBC and LLPC compartments following primary DENV2 inflection. These results provide enhanced resolution of the temporal and specificity of the B cell compartment in viral infection and serve as framework for evaluation of B cell responses in challenge models. FUNDING This study was supported by the Bill and Melinda Gates Foundation and the National Institutes of Health.
Collapse
Affiliation(s)
- Usha K Nivarthi
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Huy A Tu
- Department of Microbiology and Molecular Genetics, Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA; Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, USA
| | - Matthew J Delacruz
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Jesica Swanstrom
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Bhumi Patel
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Anna P Durbin
- Department of International Health, Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Stephen S Whitehead
- Laboratory of Infectious Diseases, NIAID, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kristen K Pierce
- Department of Microbiology and Molecular Genetics, Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Beth D Kirkpatrick
- Department of Microbiology and Molecular Genetics, Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Ralph S Baric
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA
| | - Ngan Nguyen
- Atreca, Inc. Redwood City, California 94063, USA
| | | | - Aravinda M de Silva
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, USA.
| | - Sean A Diehl
- Department of Microbiology and Molecular Genetics, Vaccine Testing Center, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA.
| |
Collapse
|
40
|
Kongpachith S, Lingampalli N, Ju CH, Blum LK, Lu DR, Elliott SE, Mao R, Robinson WH. Affinity Maturation of the Anti-Citrullinated Protein Antibody Paratope Drives Epitope Spreading and Polyreactivity in Rheumatoid Arthritis. Arthritis Rheumatol 2019; 71:507-517. [PMID: 30811898 DOI: 10.1002/art.40760] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/11/2018] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Anti-citrullinated protein antibodies (ACPAs) are a hallmark of rheumatoid arthritis (RA). While epitope spreading of the serum ACPA response is believed to contribute to RA pathogenesis, little is understood regarding how this phenomenon occurs. This study was undertaken to analyze the antibody repertoires of individuals with RA to gain insight into the mechanisms leading to epitope spreading of the serum ACPA response in RA. METHODS Plasmablasts from the blood of 6 RA patients were stained with citrullinated peptide tetramers to identify ACPA-producing B cells by flow cytometry. Plasmablasts were single-cell sorted and sequenced to obtain antibody repertoires. Sixty-nine antibodies were recombinantly expressed, and their anticitrulline reactivities were characterized using a cyclic citrullinated peptide enzyme-linked immuosorbent assay and synovial antigen arrays. Thirty-six mutated antibodies designed either to represent ancestral antibodies or to test paratope residues critical for binding, as determined from molecular modeling studies, were also tested for anticitrulline reactivities. RESULTS Clonally related monoclonal ACPAs and their shared ancestral antibodies each exhibited differential reactivity against citrullinated antigens. Molecular modeling identified residues within the complementarity-determining region loops and framework regions predicted to be important for citrullinated antigen binding. Affinity maturation resulted in mutations of these key residues, which conferred binding to different citrullinated epitopes and/or increased polyreactivity to citrullinated epitopes. CONCLUSION These results demonstrate that the different somatic hypermutations accumulated by clonally related B cells during affinity maturation alter the antibody paratope to mediate epitope spreading and polyreactivity of the ACPA response in RA, suggesting that these may be key properties that likely contribute to the pathogenicity of ACPAs.
Collapse
Affiliation(s)
- Sarah Kongpachith
- Stanford University, Stanford, California, and VA Palo Alto Health Care System, Palo Alto, California
| | - Nithya Lingampalli
- Stanford University, Stanford, California, and VA Palo Alto Health Care System, Palo Alto, California
| | - Chia-Hsin Ju
- Stanford University, Stanford, California, and VA Palo Alto Health Care System, Palo Alto, California
| | - Lisa K Blum
- Stanford University, Stanford, California, and VA Palo Alto Health Care System, Palo Alto, California
| | - Daniel R Lu
- Stanford University, Stanford, California, and VA Palo Alto Health Care System, Palo Alto, California
| | - Serra E Elliott
- Stanford University, Stanford, California, and VA Palo Alto Health Care System, Palo Alto, California
| | - Rong Mao
- Stanford University, Stanford, California, and VA Palo Alto Health Care System, Palo Alto, California
| | - William H Robinson
- Stanford University, Stanford, California, and VA Palo Alto Health Care System, Palo Alto, California
| |
Collapse
|
41
|
Pezeshki A, Ovsyannikova IG, McKinney BA, Poland GA, Kennedy RB. The role of systems biology approaches in determining molecular signatures for the development of more effective vaccines. Expert Rev Vaccines 2019; 18:253-267. [PMID: 30700167 DOI: 10.1080/14760584.2019.1575208] [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] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Emerging infectious diseases are a major threat to public health, and while vaccines have proven to be one of the most effective preventive measures for infectious diseases, we still do not have safe and effective vaccines against many human pathogens, and emerging diseases continually pose new threats. The purpose of this review is to discuss how the creation of vaccines for these new threats has been hindered by limitations in the current approach to vaccine development. Recent advances in high-throughput technologies have enabled scientists to apply systems biology approaches to collect and integrate increasingly large datasets that capture comprehensive biological changes induced by vaccines, and then decipher the complex immune response to those vaccines. AREAS COVERED This review covers advances in these technologies and recent publications that describe systems biology approaches to understanding vaccine immune responses and to understanding the rational design of new vaccine candidates. EXPERT OPINION Systems biology approaches to vaccine development provide novel information regarding both the immune response and the underlying mechanisms and can inform vaccine development.
Collapse
Affiliation(s)
| | | | - Brett A McKinney
- b Department of Mathematics , University of Tulsa , Tulsa , OK , USA.,c Tandy School of Computer Science , University of Tulsa , Tulsa , OK , USA
| | - Gregory A Poland
- a Mayo Vaccine Research Group , Mayo Clinic , Rochester , MN , USA
| | | |
Collapse
|
42
|
Shah HB, Smith K, Wren JD, Webb CF, Ballard JD, Bourn RL, James JA, Lang ML. Insights From Analysis of Human Antigen-Specific Memory B Cell Repertoires. Front Immunol 2019; 9:3064. [PMID: 30697210 PMCID: PMC6340933 DOI: 10.3389/fimmu.2018.03064] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 12/11/2018] [Indexed: 12/17/2022] Open
Abstract
Memory B cells that are generated during an infection or following vaccination act as sentinels to guard against future infections. Upon repeat antigen exposure memory B cells differentiate into new antibody-secreting plasma cells to provide rapid and sustained protection. Some pathogens evade or suppress the humoral immune system, or induce memory B cells with a diminished ability to differentiate into new plasma cells. This leaves the host vulnerable to chronic or recurrent infections. Single cell approaches coupled with next generation antibody gene sequencing facilitate a detailed analysis of the pathogen-specific memory B cell repertoire. Monoclonal antibodies that are generated from antibody gene sequences allow a functional analysis of the repertoire. This review discusses what has been learned thus far from analysis of diverse pathogen-specific memory B cell compartments and describes major differences in their repertoires. Such information may illuminate ways to advance the goal of improving vaccine and therapeutic antibody design.
Collapse
Affiliation(s)
- Hemangi B Shah
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Kenneth Smith
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Jonathan D Wren
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States.,Department of Biochemistry and Molecular Biology and Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Carol F Webb
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Division of Rheumatology, Immunology and Allergy, Department of Cell Biology and Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jimmy D Ballard
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Rebecka L Bourn
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Judith A James
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States.,Department of Medicine and Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Mark L Lang
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
43
|
Carmona-Rivera C, Bicker KL, Thompson PR, Buckner JH, Robinson WH, Fox DA, Kaplan MJ. Response to comment on "Synovial fibroblast-neutrophil interactions promote pathogenic adaptive immunity in rheumatoid arthritis". Sci Immunol 2018; 3:3/21/eaar3701. [PMID: 29602804 DOI: 10.1126/sciimmunol.aar3701] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/21/2018] [Indexed: 12/31/2022]
Abstract
The citrullinome cargo in neutrophil extracellular traps varies according to disease condition and stimulation conditions.
Collapse
Affiliation(s)
- Carmelo Carmona-Rivera
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kevin L Bicker
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, TN 37132, USA
| | - Paul R Thompson
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jane H Buckner
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - William H Robinson
- VA Palo Alto Health Care System, Palo Alto, CA 94304, USA.,Division of Rheumatology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David A Fox
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
44
|
Elliott SE, Kongpachith S, Lingampalli N, Adamska JZ, Cannon BJ, Mao R, Blum LK, Robinson WH. Affinity Maturation Drives Epitope Spreading and Generation of Proinflammatory Anti-Citrullinated Protein Antibodies in Rheumatoid Arthritis. Arthritis Rheumatol 2018; 70:1946-1958. [PMID: 29927104 PMCID: PMC6261684 DOI: 10.1002/art.40587] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 06/12/2018] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is characterized by the presence of anti-citrullinated protein antibodies (ACPAs); nevertheless, the origin, specificity, and functional properties of ACPAs remain poorly understood. The aim of this study was to characterize the evolution of ACPAs by sequencing the plasmablast antibody repertoire at serial time points in patients with established RA. METHODS Blood samples were obtained at up to 4 serial time points from 8 individuals with established RA who were positive for ACPAs by the anti-cyclic citrullinated peptide test. CD19+CD3-IgD-CD14-CD20-CD27+CD38++ plasmablasts were isolated by single-cell sorting and costained with citrullinated peptide tetramers to identify ACPA-expressing plasmablasts. Cell-specific oligonucleotide barcodes were utilized, followed by large-scale sequencing and bioinformatics analysis, to obtain error-corrected, paired heavy- and light-chain antibody gene sequences for each B cell. RESULTS Bioinformatics analysis revealed 170 persistent plasmablast lineages in the RA blood, of which 19% included multiple isotypes. Among IgG- and IgA-expressing plasmablasts, significantly more IgA-expressing than IgG-expressing persistent lineages were observed (P < 0.01). Shared complementarity-determining region 3 sequence motifs were identified across subjects. A subset of the plasmablast lineages included members derived from later time points with divergent somatic hypermutations that encoded antibodies that bind an expanded set of citrullinated antigens. Furthermore, these recombinant, differentially mutated plasmablast antibodies formed immune complexes that stimulated higher macrophage production of tumor necrosis factor (TNF) compared to antibodies representing earlier time point-derived lineage members that were less mutated. CONCLUSION These findings demonstrate that established RA is characterized by a persistent IgA ACPA response that exhibits ongoing affinity maturation. This observation suggests the presence of a persistent mucosal antigen that continually promotes the production of IgA plasmablasts and their affinity maturation and epitope spreading, thus leading to the generation of ACPAs that bind additional citrullinated antigens and more potently stimulate macrophage production of TNF.
Collapse
Affiliation(s)
- Serra E. Elliott
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA; VA Palo Alto Health Care System, Palo Alto, CA
| | - Sarah Kongpachith
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA; VA Palo Alto Health Care System, Palo Alto, CA
| | - Nithya Lingampalli
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA; VA Palo Alto Health Care System, Palo Alto, CA
| | - Julia Z. Adamska
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA; VA Palo Alto Health Care System, Palo Alto, CA
| | - Bryan J. Cannon
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA; VA Palo Alto Health Care System, Palo Alto, CA
| | - Rong Mao
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA; VA Palo Alto Health Care System, Palo Alto, CA
| | - Lisa K. Blum
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA; VA Palo Alto Health Care System, Palo Alto, CA
| | - William H. Robinson
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA; VA Palo Alto Health Care System, Palo Alto, CA
| |
Collapse
|
45
|
Lu DR, McDavid AN, Kongpachith S, Lingampalli N, Glanville J, Ju CH, Gottardo R, Robinson WH. T Cell-Dependent Affinity Maturation and Innate Immune Pathways Differentially Drive Autoreactive B Cell Responses in Rheumatoid Arthritis. Arthritis Rheumatol 2018; 70:1732-1744. [PMID: 29855173 PMCID: PMC6203609 DOI: 10.1002/art.40578] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/29/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is characterized by the activation of B cells that produce anti-citrullinated protein antibodies (ACPAs) and rheumatoid factors (RFs), but the mechanisms by which tolerance is broken in these B cells remain incompletely understood. We undertook this study to investigate whether ACPA+ and RF+ B cells break tolerance through distinct molecular mechanisms. METHODS We developed antigen-tetramers to isolate ACPA+ and RF+ B cells and performed single-cell RNA sequencing on 2,349 B cells from 6 RA patients and 1 healthy donor to analyze their immunoglobulin repertoires and transcriptional programs. Prominent immunoglobulins were expressed as monoclonal antibodies and tested for autoantigen reactivity. RESULTS ACPA+ and RF+ B cells were enriched in the peripheral blood of RA patients relative to healthy controls. Characterization of patient-derived monoclonal antibodies confirmed ACPA and RF targeting of tetramer-specific B cells at both antigen-inexperienced and affinity-matured B cell stages. ACPA+ B cells used more class-switched isotypes and exhibited more somatic hypermutations relative to RF+ B cells, and these differences were accompanied by down-regulation of CD72 and up-regulation of genes that promote class-switching and T cell-dependent responses. In contrast, RF+ B cells expressed transcriptional programs that stimulate rapid memory reactivation through multiple innate immune pathways. Coexpression analysis revealed that ACPA+ and RF+ B cell-enriched genes belong to distinct transcriptional regulatory networks. CONCLUSION Our findings suggest that ACPA+ and RF+ B cells are imprinted with distinct transcriptional programs, which suggests that these autoantibodies associated with increased inflammation in RA arise from 2 different molecular mechanisms.
Collapse
Affiliation(s)
- Daniel R. Lu
- Stanford Immunology Program, Stanford University, Stanford, CA
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA
- VA Palo Alto Health Care System, Palo Alto, CA
| | | | - Sarah Kongpachith
- Stanford Immunology Program, Stanford University, Stanford, CA
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA
- VA Palo Alto Health Care System, Palo Alto, CA
| | - Nithya Lingampalli
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA
- VA Palo Alto Health Care System, Palo Alto, CA
| | - Jacob Glanville
- Stanford Immunology Program, Stanford University, Stanford, CA
| | - Chia-Hsin Ju
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA
- VA Palo Alto Health Care System, Palo Alto, CA
| | | | - William H. Robinson
- Stanford Immunology Program, Stanford University, Stanford, CA
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA
- VA Palo Alto Health Care System, Palo Alto, CA
| |
Collapse
|
46
|
Dam EM, Maier AC, Hocking AM, Carlin J, Ng B, Buckner JH. Increased Binding of Specificity Protein 1 to the IL21R Promoter in B Cells Results in Enhanced B Cell Responses in Rheumatoid Arthritis. Front Immunol 2018; 9:1978. [PMID: 30233580 PMCID: PMC6134023 DOI: 10.3389/fimmu.2018.01978] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 08/13/2018] [Indexed: 01/09/2023] Open
Abstract
B cells are implicated in rheumatoid arthritis (RA) based on the presence of autoantibodies and the therapeutic response to B cell depletion. IL-21 has a significant role in B cell development and function. Here we assess B cell responses to IL-21 and the mechanisms responsible for altered IL-21R expression in RA. Flow cytometry of PBMC and cultured B cells was used to quantify protein and mRNA levels of IL-21R, IL-21 signaling through pSTAT3, specificity protein 1 (SP1) and to determine cytokine production (IL-6) and maturation status of B cells in RA and healthy control subjects. SP1 binding to the IL21R promoter region in B cells was assessed with ChIP-qPCR. We demonstrate an increase in IL-21R expression in total and memory B cells from RA subjects, which correlated with responsiveness to IL-21 stimulation. Stimulation of naïve RA B cells with IL-21 and CD40L resulted in an increase in differentiation into plasmablasts and an increase in IL-6 production in comparison to healthy controls, which was dose dependent on IL-21 stimulation. IL-21R expression on memory B cells in RA synovial fluid was comparable to peripheral blood making our study pertinent to understanding B cell responses in the joint and site of inflammation. We identified an increase in SP1 protein and mRNA in RA B cells and demonstrate an increase in binding of SP1 to the IL21R promoter region, which suggests a mechanism by which IL-21R expression is enhanced on B cells in RA. Taken together, our results indicate a mechanism by which IL-21 enhances B cell development and function in RA through an SP1 mediated increase in IL-21R expression on B cells.
Collapse
Affiliation(s)
- Elizabeth M Dam
- Translational Research Program, Benaroya Research Institute, Seattle, WA, United States
| | - Alison C Maier
- Translational Research Program, Benaroya Research Institute, Seattle, WA, United States
| | - Anne M Hocking
- Translational Research Program, Benaroya Research Institute, Seattle, WA, United States
| | - Jeffrey Carlin
- Division of Rheumatology, Virginia Mason Medical Center, Seattle, WA, United States
| | - Bernard Ng
- Rheumatology Section, VA Puget Sound Health Care System, Seattle, WA, United States,Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Jane H Buckner
- Translational Research Program, Benaroya Research Institute, Seattle, WA, United States
| |
Collapse
|
47
|
Holers VM, Demoruelle MK, Kuhn KA, Buckner JH, Robinson WH, Okamoto Y, Norris JM, Deane KD. Rheumatoid arthritis and the mucosal origins hypothesis: protection turns to destruction. Nat Rev Rheumatol 2018; 14:542-557. [PMID: 30111803 PMCID: PMC6704378 DOI: 10.1038/s41584-018-0070-0] [Citation(s) in RCA: 225] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Individuals at high risk of developing seropositive rheumatoid arthritis (RA) can be identified for translational research and disease prevention studies through the presence of highly informative and predictive patterns of RA-related autoantibodies, especially anti-citrullinated protein antibodies (ACPAs), in the serum. In serologically positive individuals without arthritis, designated ACPA positive at risk, the presence of mucosal inflammatory processes associated with the presence of local ACPA production has been demonstrated. In other at-risk populations, local RA-related autoantibody production is present even in the absence of serum autoantibodies. Additionally, a proportion of at-risk individuals exhibit local mucosal ACPA production in the lung, as well as radiographic small-airway disease, sputum hypercellularity and increased neutrophil extracellular trap formation. Other mucosal sites in at-risk individuals also exhibit autoantibody production, inflammation and/or evidence of dysbiosis. As the proportion of individuals who exhibit such localized inflammation-associated ACPA production is substantially higher than the likelihood of an individual developing future RA, this finding raises the hypothesis that mucosal ACPAs have biologically relevant protective roles. Identifying the mechanisms that drive both the generation and loss of externally focused mucosal ACPA production and promote systemic autoantibody expression and ultimately arthritis development should provide insights into new therapeutic approaches to prevent RA.
Collapse
Affiliation(s)
- V Michael Holers
- Division of Rheumatology, University of Colorado-Denver, Aurora, CO, USA.
| | | | - Kristine A Kuhn
- Division of Rheumatology, University of Colorado-Denver, Aurora, CO, USA
| | | | - William H Robinson
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, USA
| | - Yuko Okamoto
- Division of Rheumatology, University of Colorado-Denver, Aurora, CO, USA
| | - Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - Kevin D Deane
- Division of Rheumatology, University of Colorado-Denver, Aurora, CO, USA
| |
Collapse
|
48
|
Blum LK, Adamska JZ, Martin DS, Rebman AW, Elliott SE, Cao RRL, Embers ME, Aucott JN, Soloski MJ, Robinson WH. Robust B Cell Responses Predict Rapid Resolution of Lyme Disease. Front Immunol 2018; 9:1634. [PMID: 30072990 PMCID: PMC6060717 DOI: 10.3389/fimmu.2018.01634] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/03/2018] [Indexed: 12/11/2022] Open
Abstract
Lyme disease (Borrelia burgdorferi infection) is increasingly recognized as a significant source of morbidity worldwide. Here, we show that blood plasmablasts and CD27− memory B cells are elevated in untreated Lyme disease, with higher plasmablast levels associated with more rapid resolution of clinical symptoms. Stronger serum reactivity to surface proteins and peptides from B. burgdorferi was also associated with faster resolution of clinical symptoms. Through molecular identifier-enabled antibody heavy-chain sequencing of bulk B cells and single-cell paired-chain antibody sequencing of blood plasmablasts, we characterized immunoglobulin gene usage patterns specific to B. burgdorferi infection. Recombinantly expressed antibodies from expanded lineages bound B. burgdorferi antigens, confirming that these clones are driven by the infection. Furthermore, recombinant sequence-derived antibodies were functional, inhibiting growth of B. burgdorferi in vitro. Elevations and clonal expansion of blood plasmablasts were associated with rapid return to health, while poor plasmablast responses were associated with a longer duration of symptoms following treatment. Plasmablasts induced by B. burgdorferi infection showed preferential antibody gene segment usage, while bulk sequencing of total B cells revealed convergent CDR3 motifs specific to B. burgdorferi-infected patients. Our results show that robust plasmablast responses encoding Bb-static antibodies are associated with more rapid resolution of Lyme disease, and these antibodies could provide the basis for next-generation therapeutics for Lyme disease.
Collapse
Affiliation(s)
- Lisa K Blum
- Stanford University School of Medicine, Stanford, CA, United States.,VA Palo Alto Healthcare System, Palo Alto, CA, United States
| | - Julia Z Adamska
- Stanford University School of Medicine, Stanford, CA, United States.,VA Palo Alto Healthcare System, Palo Alto, CA, United States
| | - Dale S Martin
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States
| | - Alison W Rebman
- Lyme Disease Research Center, Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Serra E Elliott
- Stanford University School of Medicine, Stanford, CA, United States.,VA Palo Alto Healthcare System, Palo Alto, CA, United States
| | - Richard R L Cao
- Stanford University School of Medicine, Stanford, CA, United States
| | - Monica E Embers
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University Health Sciences Center, Covington, LA, United States
| | - John N Aucott
- Lyme Disease Research Center, Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mark J Soloski
- Lyme Disease Research Center, Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - William H Robinson
- Stanford University School of Medicine, Stanford, CA, United States.,VA Palo Alto Healthcare System, Palo Alto, CA, United States
| |
Collapse
|
49
|
Bashford-Rogers RJM, Smith KGC, Thomas DC. Antibody repertoire analysis in polygenic autoimmune diseases. Immunology 2018; 155:3-17. [PMID: 29574826 PMCID: PMC6099162 DOI: 10.1111/imm.12927] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/01/2018] [Accepted: 03/06/2018] [Indexed: 12/18/2022] Open
Abstract
High-throughput sequencing of the DNA/RNA encoding antibody heavy- and light-chains is rapidly transforming the field of adaptive immunity. It can address key questions, including: (i) how the B-cell repertoire differs in health and disease; and (ii) if it does differ, the point(s) in B-cell development at which this occurs. The advent of technologies, such as whole-genome sequencing, offers the chance to link abnormalities in the B-cell antibody repertoire to specific genomic variants and polymorphisms. Here, we discuss the current research using B-cell antibody repertoire sequencing in three polygenic autoimmune diseases where there is good evidence for a pathological role for B-cells, namely systemic lupus erythematosus, multiple sclerosis and rheumatoid arthritis. These autoimmune diseases exhibit significantly skewed B-cell receptor repertoires compared with healthy controls. Interestingly, some common repertoire defects are shared between diseases, such as elevated IGHV4-34 gene usage. B-cell clones have effectively been characterized and tracked between different tissues and blood in autoimmune disease. It has been hypothesized that these differences may signify differences in B-cell tolerance; however, the mechanisms and implications of these defects are not clear.
Collapse
Affiliation(s)
| | | | - David C Thomas
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
50
|
Upadhyay AA, Kauffman RC, Wolabaugh AN, Cho A, Patel NB, Reiss SM, Havenar-Daughton C, Dawoud RA, Tharp GK, Sanz I, Pulendran B, Crotty S, Lee FEH, Wrammert J, Bosinger SE. BALDR: a computational pipeline for paired heavy and light chain immunoglobulin reconstruction in single-cell RNA-seq data. Genome Med 2018; 10:20. [PMID: 29558968 PMCID: PMC5859752 DOI: 10.1186/s13073-018-0528-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 02/23/2018] [Indexed: 01/21/2023] Open
Abstract
B cells play a critical role in the immune response by producing antibodies, which display remarkable diversity. Here we describe a bioinformatic pipeline, BALDR (BCR Assignment of Lineage using De novo Reconstruction) that accurately reconstructs the paired heavy and light chain immunoglobulin gene sequences from Illumina single-cell RNA-seq data. BALDR was accurate for clonotype identification in human and rhesus macaque influenza vaccine and simian immunodeficiency virus vaccine induced vaccine-induced plasmablasts and naïve and antigen-specific memory B cells. BALDR enables matching of clonotype identity with single-cell transcriptional information in B cell lineages and will have broad application in the fields of vaccines, human immunodeficiency virus broadly neutralizing antibody development, and cancer. BALDR is available at https://github.com/BosingerLab/BALDR.
Collapse
Affiliation(s)
- Amit A Upadhyay
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Robert C Kauffman
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Amber N Wolabaugh
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Alice Cho
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Nirav B Patel
- Yerkes NHP Genomics Core Laboratory, Yerkes National Primate Research Center, 954 Gatewood Rd, Atlanta, GA, 30329, USA
| | - Samantha M Reiss
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA.,Scripps Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery (CHAVI-ID), La Jolla, CA, USA
| | - Colin Havenar-Daughton
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA.,Scripps Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery (CHAVI-ID), La Jolla, CA, USA
| | - Reem A Dawoud
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Gregory K Tharp
- Yerkes NHP Genomics Core Laboratory, Yerkes National Primate Research Center, 954 Gatewood Rd, Atlanta, GA, 30329, USA
| | - Iñaki Sanz
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA.,Division of Rheumatology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA.,Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Shane Crotty
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA.,Scripps Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery (CHAVI-ID), La Jolla, CA, USA.,Division of Infectious Diseases, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - F Eun-Hyung Lee
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA.,Divisions of Pulmonary, Allergy and Critical Care Medicine, Emory University, Atlanta, GA, USA
| | - Jens Wrammert
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Steven E Bosinger
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Atlanta, GA, USA. .,Yerkes NHP Genomics Core Laboratory, Yerkes National Primate Research Center, 954 Gatewood Rd, Atlanta, GA, 30329, USA. .,Department of Pathology & Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, USA.
| |
Collapse
|