1
|
Li R, Bao K, Liu C, Ma X, Hua Z, Zhu P, Hou B. Competition propels, rather than limits, the success of low-affinity B cells in the germinal center response. Cell Rep 2025; 44:115334. [PMID: 39955776 DOI: 10.1016/j.celrep.2025.115334] [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: 06/18/2024] [Revised: 12/01/2024] [Accepted: 01/29/2025] [Indexed: 02/18/2025] Open
Abstract
The germinal center (GC) sets an environment where antigen-specific B cells are compelled to continuously increase their affinity to compete for the antigen and obtain Tfh help for survival and propagation. Previous studies indicated that low-affinity B cells are disadvantaged in the presence of high-affinity ones, suggesting that competition may lead to the elimination of low-affinity B cells and their descendants. However, using a multivalent virus-mimicking antigen, our study demonstrates that low-affinity B cells not only successfully participate in GC responses alongside high-affinity B cells but also undergo accelerated affinity maturation under the more stringent competition. Furthermore, our cryo-electron-microscopy-based structural analysis reveals that both low-affinity and high-affinity B cells compete for the same antigenic epitope. Although the applicability of this idealized GC competition to true pathogen-induced responses remains uncertain, this change in perspective on the role of competition in low-affinity B cell evolution provides valuable insights for vaccine development.
Collapse
Affiliation(s)
- Runhan Li
- State Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Keyan Bao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Can Liu
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuejing Ma
- State Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhaolin Hua
- State Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Ping Zhu
- State Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Baidong Hou
- State Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
2
|
Guo W, Wei M, Li Y, Xu J, Zang J, Chen Y, Chen L. Mechanisms of urate transport and uricosuric drugs inhibition in human URAT1. Nat Commun 2025; 16:1512. [PMID: 39929841 PMCID: PMC11811179 DOI: 10.1038/s41467-025-56843-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 02/02/2025] [Indexed: 02/13/2025] Open
Abstract
High urate levels in circulation lead to the accumulation of urate crystals in joints and ultimately inflammation and gout. The reabsorption process of urate in the kidney by the urate transporter URAT1 plays a pivotal role in controlling serum urate levels. Pharmacological inhibition of URAT1 by uricosuric drugs is a valid strategy for gout management. Despite the clinical significance of URAT1, its structural mechanism and dynamics remain incompletely understood. Here, we report the structures of human URAT1 (hURAT1) in complex with substrate urate or inhibitors benzbromarone and verinurad at resolution ranges from 3.0 to 3.3 Å. We observe urate in the central substrate-binding site of hURAT1 in the outward-facing conformation and urate is wrapped in the center of hURAT1 by five phenylalanines and coordinated by two positively charged residues on each side. Uricosuric compounds benzbromarone and verinurad occupy the urate-binding site of hURAT1 in the inward-facing conformation. Structural comparison between different conformations of hURAT1 reveals the rocker-switch-like mechanism for urate transport. Benzbromarone and verinurad exert their inhibitory effect by blocking not only the binding of urate but also the structural isomerization of hURAT1.
Collapse
Affiliation(s)
- Wenjun Guo
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking. University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- National Biomedical Imaging Center, Peking University, Beijing, 100871, China
| | - Miao Wei
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking. University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, 100871, China
| | - Yunfeng Li
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Jiaxuan Xu
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking. University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, 100871, China
| | - Jiahe Zang
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking. University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, 100871, China
| | - Yuezhou Chen
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Lei Chen
- State Key Laboratory of Membrane Biology, College of Future Technology, Institute of Molecular Medicine, Peking. University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, 100871, China.
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
- National Biomedical Imaging Center, Peking University, Beijing, 100871, China.
| |
Collapse
|
3
|
Termote M, Marques RC, Hyllner E, Guryleva MV, Henskens M, Brutscher A, Baken IJL, Dopico XC, Gasull AD, Murrell B, Stamatatos L, Westerberg LS, Dosenovic P. Antigen affinity and site of immunization dictate B cell recall responses. Cell Rep 2025; 44:115221. [PMID: 39817910 DOI: 10.1016/j.celrep.2024.115221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 11/29/2024] [Accepted: 12/26/2024] [Indexed: 01/18/2025] Open
Abstract
Protective antibodies against HIV-1 require unusually high levels of somatic mutations introduced in germinal centers (GCs). To achieve this, a sequential vaccination approach was proposed. Using HIV-1 antibody knockin mice with fate-mapping genes, we examined if antigen affinity affects the outcome of B cell recall responses. Compared to a high-affinity boost, a low-affinity boost resulted in decreased numbers of memory-derived B cells in secondary GCs but with higher average levels of somatic mutations, indicating an affinity threshold for memory B cells to enter GCs. Furthermore, upon boosting local lymph nodes (LNs), the composition of primary GCs was modified in an antigen-affinity-dependent manner to constitute less somatically mutated B cells. Our results demonstrate that antigen affinity and location of the boost affect the outcome of the B cell recall response. These results can help guide the design of vaccine immunogens aiming to selectively engage specific B cell clones for further diversification.
Collapse
Affiliation(s)
- Manon Termote
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Rafael C Marques
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Erik Hyllner
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Mariia V Guryleva
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Mirthe Henskens
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Andreas Brutscher
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Isabel J L Baken
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Xaquin Castro Dopico
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Adria Dalmau Gasull
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Ben Murrell
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Leonidas Stamatatos
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Lisa S Westerberg
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Pia Dosenovic
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden.
| |
Collapse
|
4
|
Rollenske T, Murugan R, Wardemann H, Busse CE. Expression Cloning of Antibodies from Single Human B Cells. Methods Mol Biol 2025; 2865:103-124. [PMID: 39424722 DOI: 10.1007/978-1-0716-4188-0_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
The majority of lymphomas originate from B cells at the germinal center stage. Preferential selection of B-cell clones by a limited set of antigens has been suggested to drive lymphoma development. While recent studies in B-cell chronic lymphocytic leukemia (CLL) have shown that self-reactive B-cell receptors (BCR) can generate cell-autonomous signaling and proliferation, our knowledge about the role of BCRs for the development or survival of other lymphomas remains limited. Here, we describe a strategy to characterize the antibody reactivity of human B cells. The approach allows for unbiased characterization of the human antibody repertoire on single-cell level through the generation of recombinant monoclonal antibodies from primary human B cells of defined origin. This protocol offers a detailed description of the method starting from the flow cytometric isolation of single human B cells to the RT-PCR-based amplification of the expressed immunoglobulin (Ig) transcripts (IGH, IGK, and IGL) and their subsequent cloning into expression vectors for the in vitro production of recombinant monoclonal antibodies. The strategy may be used to obtain information about the clonal evolution of B-cell lymphomas by single-cell sequencing of Ig transcripts and on the antibody reactivity of human lymphoma B cells.
Collapse
Affiliation(s)
- Tim Rollenske
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Rheinische Friedrich Wilhelm University, Bonn, Germany
| | - Rajagopal Murugan
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Hedda Wardemann
- Division of B Cell Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian E Busse
- Division of B Cell Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
5
|
Ado S, Dong C, Attaf N, Moussa M, Carrier A, Milpied P, Navarro JM. FB5P-seq-mAbs: monoclonal antibody production from FB5P-seq libraries for integrative single-cell analysis of B cells. Front Immunol 2024; 15:1505971. [PMID: 39742275 PMCID: PMC11685048 DOI: 10.3389/fimmu.2024.1505971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 11/26/2024] [Indexed: 01/03/2025] Open
Abstract
Parallel analysis of phenotype, transcriptome and antigen receptor sequence in single B cells is a useful method for tracking B cell activation and maturation during immune responses. However, in most cases, the specificity and affinity of the B cell antigen receptor cannot be inferred from its sequence. Antibody cloning and expression from single B cells is then required for functional assays. Here we propose a method that integrates FACS-based 5'-end single-cell RNA sequencing (FB5P-seq) and monoclonal antibody cloning for integrative analysis of single B cells. Starting from a cell suspension, single B cells are FACS-sorted into 96-well plates for reverse transcription, cDNA barcoding and amplification. A fraction of the single-cell cDNA is used for preparing 5'-end RNA-seq libraries that are sequenced for retrieving transcriptome-wide gene expression and paired BCR sequences. The archived cDNA of selected cells of interest is used as input for cloning heavy and light chain variable regions into antibody expression plasmid vectors. The corresponding monoclonal antibodies are produced by transient transfection of a eukaryotic producing cell line and purified for functional assays. We provide detailed step-by-step instructions and describe results obtained on ovalbumin-specific murine germinal center B cells after immunization. Our method is robust, flexible, cost-effective, and applicable to different B cell types and species. We anticipate it will be useful for mapping antigen specificity and affinity of rare B cell subsets characterized by defined gene expression and/or antigen receptor sequence.
Collapse
Affiliation(s)
- Sakina Ado
- Aix Marseille Université, CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Chuang Dong
- Aix Marseille Université, CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Noudjoud Attaf
- Aix Marseille Université, CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Myriam Moussa
- Aix Marseille Université, CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Agathe Carrier
- Aix Marseille Université, CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Marseille, France
- Paris-Saclay University, Inserm, Gustave Roussy, Tumour Immunology and Anti-Cancer Immunotherapy, Villejuif, France
| | - Pierre Milpied
- Aix Marseille Université, CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| | - Jean-Marc Navarro
- Aix Marseille Université, CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Marseille, France
| |
Collapse
|
6
|
Lopatto EDB, Santiago-Borges JM, Sanick DA, Malladi SK, Azimzadeh PN, Timm MW, Fox IF, Schmitz AJ, Turner JS, Ahmed SS, Ortinau L, Gualberto NC, Pinkner JS, Dodson KW, Ellebedy AH, Kau AL, Hultgren SJ. Monoclonal antibodies targeting the FimH adhesin protect against uropathogenic E. coli UTI. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.10.627638. [PMID: 39713358 PMCID: PMC11661314 DOI: 10.1101/2024.12.10.627638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
As antimicrobial resistance increases, urinary tract infections (UTIs) are expected to pose an increased burden in morbidity and expense on the healthcare system, increasing the need for alternative antibiotic-sparing treatments. Most UTIs are caused by uropathogenic Escherichia coli (UPEC), while Klebsiella pneumoniae causes a significant portion of non-UPEC UTIs. Both bacteria express type 1 pili tipped with the mannose-binding FimH adhesin critical for UTI pathogenesis. We generated and biochemically characterized 33 murine monoclonal antibodies (mAbs) to FimH. Two mAbs protected mice from E. coli UTI. Mechanistically, we show that this protection is Fc-independent and mediated by the ability of these mAbs to sterically block FimH function. Our data reveals that FimH mAbs hold promise as an antibiotic-sparing treatment strategy.
Collapse
Affiliation(s)
- Edward D. B. Lopatto
- Department of Molecular Microbiology, Washington University in St Louis, St Louis, MO, U.S.A
- Center for Women’s Infectious Disease Research, Washington University in St Louis, St Louis, MO, U.S.A
| | - Jesús M. Santiago-Borges
- Department of Molecular Microbiology, Washington University in St Louis, St Louis, MO, U.S.A
- Center for Women’s Infectious Disease Research, Washington University in St Louis, St Louis, MO, U.S.A
- Division of Allergy and Immunology, Department of Medicine, Washington University in St Louis, St Louis, MO, U.S.A
| | - Denise A. Sanick
- Department of Molecular Microbiology, Washington University in St Louis, St Louis, MO, U.S.A
- Center for Women’s Infectious Disease Research, Washington University in St Louis, St Louis, MO, U.S.A
| | - Sameer Kumar Malladi
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, U.S.A
| | - Philippe N. Azimzadeh
- Department of Molecular Microbiology, Washington University in St Louis, St Louis, MO, U.S.A
- Center for Women’s Infectious Disease Research, Washington University in St Louis, St Louis, MO, U.S.A
| | - Morgan W. Timm
- Department of Molecular Microbiology, Washington University in St Louis, St Louis, MO, U.S.A
- Center for Women’s Infectious Disease Research, Washington University in St Louis, St Louis, MO, U.S.A
| | - Isabella F. Fox
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, U.S.A
| | - Aaron J. Schmitz
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, U.S.A
| | - Jackson S. Turner
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, U.S.A
| | - Shaza Sayed Ahmed
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, U.S.A
| | - Lillian Ortinau
- Department of Molecular Microbiology, Washington University in St Louis, St Louis, MO, U.S.A
- Center for Women’s Infectious Disease Research, Washington University in St Louis, St Louis, MO, U.S.A
- Division of Allergy and Immunology, Department of Medicine, Washington University in St Louis, St Louis, MO, U.S.A
| | - Nathaniel C. Gualberto
- Department of Molecular Microbiology, Washington University in St Louis, St Louis, MO, U.S.A
- Center for Women’s Infectious Disease Research, Washington University in St Louis, St Louis, MO, U.S.A
| | - Jerome S. Pinkner
- Department of Molecular Microbiology, Washington University in St Louis, St Louis, MO, U.S.A
- Center for Women’s Infectious Disease Research, Washington University in St Louis, St Louis, MO, U.S.A
| | - Karen W. Dodson
- Department of Molecular Microbiology, Washington University in St Louis, St Louis, MO, U.S.A
- Center for Women’s Infectious Disease Research, Washington University in St Louis, St Louis, MO, U.S.A
| | - Ali H. Ellebedy
- Department of Molecular Microbiology, Washington University in St Louis, St Louis, MO, U.S.A
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, U.S.A
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO, U.S.A
| | - Andrew L. Kau
- Department of Molecular Microbiology, Washington University in St Louis, St Louis, MO, U.S.A
- Center for Women’s Infectious Disease Research, Washington University in St Louis, St Louis, MO, U.S.A
- Division of Allergy and Immunology, Department of Medicine, Washington University in St Louis, St Louis, MO, U.S.A
| | - Scott J. Hultgren
- Department of Molecular Microbiology, Washington University in St Louis, St Louis, MO, U.S.A
- Center for Women’s Infectious Disease Research, Washington University in St Louis, St Louis, MO, U.S.A
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO, U.S.A
| |
Collapse
|
7
|
Morgan DM, Zhang YJ, Kim JH, Murillo M, Singh S, Loschko J, Surendran N, Sekulovic O, Feng E, Shi S, Irvine DJ, Patil SU, Kanevsky I, Chorro L, Christopher Love J. Full-length single-cell BCR sequencing paired with RNA sequencing reveals convergent responses to pneumococcal vaccination. Commun Biol 2024; 7:1208. [PMID: 39341987 PMCID: PMC11438910 DOI: 10.1038/s42003-024-06823-0] [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: 06/13/2023] [Accepted: 09/02/2024] [Indexed: 10/01/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) can resolve transcriptional features from individual cells, but scRNA-seq techniques capable of resolving the variable regions of B cell receptors (BCRs) remain limited, especially from widely-used 3'-barcoded libraries. Here, we report a method that can recover paired, full-length variable region sequences of BCRs from 3'-barcoded scRNA-seq libraries. We first verify this method (B3E-seq) can produce accurate, full-length BCR sequences. We then apply this method to profile B cell responses elicited against the capsular polysaccharide of Streptococcus pneumoniae serotype 3 (ST3) by glycoconjugate vaccines in five infant rhesus macaques. We identify BCR features associated with specificity for the ST3 antigen which are present in multiple vaccinated monkeys, indicating a convergent response to vaccination. These results demonstrate the utility of our method to resolve key features of the B cell repertoire and profile antigen-specific responses elicited by vaccination.
Collapse
Affiliation(s)
- Duncan M Morgan
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Chemical Engineering, MIT, Cambridge, MA, USA
| | - Yiming J Zhang
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Biological Engineering, MIT, Cambridge, MA, USA
| | - Jin-Hwan Kim
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - MaryAnn Murillo
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - Suddham Singh
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - Jakob Loschko
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
- Deerfield Management, New York, NY, USA
| | - Naveen Surendran
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - Ognjen Sekulovic
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - Ellie Feng
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Biological Engineering, MIT, Cambridge, MA, USA
| | - Shuting Shi
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Chemical Engineering, MIT, Cambridge, MA, USA
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Department of Biological Engineering, MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Materials Science and Engineering, MIT, Cambridge, MA, USA
| | - Sarita U Patil
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Isis Kanevsky
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
| | - Laurent Chorro
- Vaccine Research and Development Pfizer Inc. Pearl River, New York, NY, USA
- Regeneron, Tarrytown, NY, USA
| | - J Christopher Love
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA.
- Department of Chemical Engineering, MIT, Cambridge, MA, USA.
| |
Collapse
|
8
|
Berndsen ZT, Akhtar M, Thapa M, Vickers TJ, Schmitz A, Torres JL, Baboo S, Kumar P, Khatoon N, Sheikh A, Hamrick M, Diedrich JK, Martinez-Bartolome S, Garrett PT, Yates JR, Turner JS, Laird RM, Poly F, Porter CK, Copps J, Ellebedy AH, Ward AB, Fleckenstein JM. Repeat modules and N-linked glycans define structure and antigenicity of a critical enterotoxigenic E. coli adhesin. PLoS Pathog 2024; 20:e1012241. [PMID: 39283948 PMCID: PMC11463764 DOI: 10.1371/journal.ppat.1012241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/09/2024] [Accepted: 08/12/2024] [Indexed: 09/25/2024] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) cause hundreds of millions of cases of infectious diarrhea annually, predominantly in children from low-middle income regions. Notably, in children, as well as volunteers challenged with ETEC, diarrheal severity is significantly increased in blood group A (bgA) individuals. EtpA, is a secreted glycoprotein adhesin that functions as a blood group A lectin to promote critical interactions between ETEC and blood group A glycans on intestinal epithelia for effective bacterial adhesion and toxin delivery. EtpA is highly immunogenic resulting in robust antibody responses following natural infection and experimental challenge of volunteers with ETEC. To understand how EtpA directs ETEC-blood group A interactions and stimulates adaptive immunity, we mutated EtpA, mapped its glycosylation by mass-spectrometry (MS), isolated polyclonal (pAbs) and monoclonal antibodies (mAbs) from vaccinated mice and ETEC-infected volunteers, and determined structures of antibody-EtpA complexes by cryo-electron microscopy. Both bgA and mAbs that inhibited EtpA-bgA interactions and ETEC adhesion, bound to the C-terminal repeat domain highlighting this region as crucial for ETEC pathogen-host interaction. MS analysis uncovered extensive and heterogeneous N-linked glycosylation of EtpA and cryo-EM structures revealed that mAbs directly engage these unique glycan containing epitopes. Finally, electron microscopy-based polyclonal epitope mapping revealed antibodies targeting numerous distinct epitopes on N and C-terminal domains, suggesting that EtpA vaccination generates responses against neutralizing and decoy regions of the molecule. Collectively, we anticipate that these data will inform our general understanding of pathogen-host glycan interactions and adaptive immunity relevant to rational vaccine subunit design.
Collapse
Affiliation(s)
- Zachary T. Berndsen
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
| | - Marjahan Akhtar
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Mahima Thapa
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, United States of America
| | - Tim J. Vickers
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Aaron Schmitz
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, United States of America
| | - Jonathan L. Torres
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
| | - Sabyasachi Baboo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Pardeep Kumar
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Nazia Khatoon
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Alaullah Sheikh
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Melissa Hamrick
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Jolene K. Diedrich
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Salvador Martinez-Bartolome
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Patrick T. Garrett
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - John R. Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Jackson S. Turner
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, United States of America
| | - Renee M. Laird
- Operationally Relevant Infections Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, United States of America
| | - Frédéric Poly
- Operationally Relevant Infections Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, United States of America
| | - Chad K. Porter
- Translational and Clinical Research Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, United States of America
| | - Jeffrey Copps
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
| | - Ali H. Ellebedy
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, United States of America
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, Missouri, United States of America
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Andrew B. Ward
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
| | - James M. Fleckenstein
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
- Operationally Relevant Infections Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, United States of America
- Medicine Service, Infectious Diseases, Veterans Affairs Health Care System, Saint Louis, Missouri, United States of America
| |
Collapse
|
9
|
Tang X, He L, Wang X, Liu S, Liu X, Shen X, Shu Y, Yang K, Zhou Q, Shan Z, Wang Y, Wu C, Jia Z, Liu T, Wang Y, Liao HX, Xia Y. Isolation of anti-tumor monoclonal antibodies targeting on MICA/B α3 domain by single B cell technology for colon cancer therapy. Heliyon 2024; 10:e35697. [PMID: 39170144 PMCID: PMC11336886 DOI: 10.1016/j.heliyon.2024.e35697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
Colon cancer (CC) is one of the most common gastrointestinal malignancies. Effectiveness of the existing therapies is limited. Immunotherapy is a promising complementary treatment approach for CC. Major histocompatibility complex class I-related protein A and B (MICA/B) are ligands for NK cells. Shedding of MICA/B from the surface of tumor cells by cleavage of MICA/B at the membrane proxial region in MICA/B α3 structural domain is one of immune evasion strategies leading to escape of cancer cells from immunosurveillance. In this study, we generated a panel of MICA/B monoclonal antibodies (mAbs) and identified one of mAbs, mAb RDM028, that had high binding affinity to MICA/B and recognized a site on MICA/B α3 structural domain that is critically important for cleavage of MICA/B. Our study has further demonstrated that RDM028 augmented the surface expression of MICA/B on HCT-116 human CC cells by inhibiting the MICA/B shedding resulting in the enhanced cyotoxicity of NK cells against HCT-116 human CC cells and mediated anti-tumor activity in nude mouse model of colon cancer. These results indicate that mAb RDM028 could be explored for developing as an effective immuno therapy against CC by targeting the MICA/B α3 domain to promot immunosurveillance mediated by MICA/B-NKG2D interaction.
Collapse
Affiliation(s)
- Xueyi Tang
- The People's Hospital of Xishuangbanna Dai Nationality Autonomous Prefecture, Xishuangbanna Dai Nationality Autonomous Prefecture, Yunnan, China
| | - Linhai He
- The People's Hospital of Xishuangbanna Dai Nationality Autonomous Prefecture, Xishuangbanna Dai Nationality Autonomous Prefecture, Yunnan, China
| | - Xiaoli Wang
- Zhuhai Trinomab Pharmaceutical Co., Ltd, Zhuhai, Guangdong, China
| | - Shuaichao Liu
- Zhuhai Trinomab Pharmaceutical Co., Ltd, Zhuhai, Guangdong, China
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiangning Liu
- Clinical Research Platform for Interdiscipline of Stomatology, The First Affiliated Hospital of Jinan University & Department of Stomatology, College of Stomatology, Jinan University, Guangzhou, China
| | - Xiaorui Shen
- The People's Hospital of Xishuangbanna Dai Nationality Autonomous Prefecture, Xishuangbanna Dai Nationality Autonomous Prefecture, Yunnan, China
| | - Yun Shu
- The People's Hospital of Xishuangbanna Dai Nationality Autonomous Prefecture, Xishuangbanna Dai Nationality Autonomous Prefecture, Yunnan, China
| | - Ke Yang
- The People's Hospital of Xishuangbanna Dai Nationality Autonomous Prefecture, Xishuangbanna Dai Nationality Autonomous Prefecture, Yunnan, China
| | - Qionghua Zhou
- The People's Hospital of Xishuangbanna Dai Nationality Autonomous Prefecture, Xishuangbanna Dai Nationality Autonomous Prefecture, Yunnan, China
| | - Zujian Shan
- The People's Hospital of Xishuangbanna Dai Nationality Autonomous Prefecture, Xishuangbanna Dai Nationality Autonomous Prefecture, Yunnan, China
| | - Yueming Wang
- Zhuhai Trinomab Pharmaceutical Co., Ltd, Zhuhai, Guangdong, China
| | - Changwen Wu
- Zhuhai Trinomab Pharmaceutical Co., Ltd, Zhuhai, Guangdong, China
| | - Zhenxing Jia
- Zhuhai Trinomab Pharmaceutical Co., Ltd, Zhuhai, Guangdong, China
| | - Tong Liu
- Zhuhai Trinomab Pharmaceutical Co., Ltd, Zhuhai, Guangdong, China
| | - Yayu Wang
- Zhuhai Trinomab Pharmaceutical Co., Ltd, Zhuhai, Guangdong, China
| | - Hua-Xin Liao
- Zhuhai Trinomab Pharmaceutical Co., Ltd, Zhuhai, Guangdong, China
| | - Yun Xia
- The People's Hospital of Xishuangbanna Dai Nationality Autonomous Prefecture, Xishuangbanna Dai Nationality Autonomous Prefecture, Yunnan, China
| |
Collapse
|
10
|
Iizuka-Koga M, Ito M, Yumoto N, Mise-Omata S, Hayakawa T, Komai K, Chikuma S, Takahashi S, Matsumoto I, Sumida T, Yoshimura A. Reconstruction of Sjögren's syndrome-like sialadenitis by a defined disease specific gut-reactive single TCR and an autoantibody. Clin Immunol 2024; 264:110258. [PMID: 38762063 DOI: 10.1016/j.clim.2024.110258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
Lymphocytes such as CD4+ T cells and B cells mainly infiltrate the salivary glands; however, the precise roles and targets of autoreactive T cells and autoantibodies in the pathogenesis of Sjögren's Syndrome (SS) remain unclear. This study was designed to clarify the role of autoreactive T cells and autoantibodies at the single-cell level involved in the development of sialadenitis. Infiltrated CD4+ T and B cells in the salivary glands of a mouse model resembling SS were single-cell-sorted, and their T cell receptor (TCR) and B cell receptor (BCR) sequences were analyzed. The predominant TCR and BCR clonotypes were reconstituted in vitro, and their pathogenicity was evaluated by transferring reconstituted TCR-expressing CD4+ T cells into Rag2-/- mice and administering recombinant IgG in vivo. The reconstitution of Th17 cells expressing TCR (#G) in Rag2-/- mice resulted in the infiltration of T cells into the salivary glands and development of sialadenitis, while an autoantibody (IgGr22) was observed to promote the proliferation of pathogenic T cells. IgGr22 specifically recognizes double-stranded RNA (dsRNA) and induces the activation of dendritic cells, thereby enhancing the expression of IFN signature and inflammatory genes. TCR#G recognizes antigens related to the gut microbiota. Antibiotic treatment severely reduces the activation of TCR#G-expressing Th17 cells and suppresses sialadenitis development. These data suggest that the anti-dsRNA antibodies and, TCR recognizing the gut microbiota involved in the development of sialadenitis like SS. Thus, our model provides a novel strategy for defining the roles of autoreactive TCR and autoantibodies in the development and pathogenesis of SS.
Collapse
Affiliation(s)
- Mana Iizuka-Koga
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Minako Ito
- Division of Allergy and Immunology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Noriko Yumoto
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Setsuko Mise-Omata
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki 2669, Noda-shi, Chiba 278-0022, Japan
| | - Taeko Hayakawa
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kyoko Komai
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shunsuke Chikuma
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Isao Matsumoto
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Takayuki Sumida
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki 2669, Noda-shi, Chiba 278-0022, Japan.
| |
Collapse
|
11
|
Alhassan AM, Shirure VS, Luo J, Nguyen BB, Rollins ZA, Shergill BS, Zhu X, Baumgarth N, George SC. A Microfluidic Strategy to Capture Antigen‐Specific High‐Affinity B Cells. ADVANCED NANOBIOMED RESEARCH 2024; 4. [DOI: 10.1002/anbr.202300101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Assessing B cell affinity to pathogen‐specific antigens prior to or following exposure could facilitate the assessment of immune status. Current standard tools to assess antigen‐specific B cell responses focus on equilibrium binding of the secreted antibody in serum. These methods are costly, time‐consuming, and assess antibody affinity under zero force. Recent findings indicate that force may influence BCR‐antigen binding interactions and thus immune status. Herein, a simple laminar flow microfluidic chamber in which the antigen (hemagglutinin of influenza A) is bound to the chamber surface to assess antigen‐specific BCR binding affinity of five hemagglutinin‐specific hybridomas from 65 to 650 pN force range is designed. The results demonstrate that both increasing shear force and bound lifetime can be used to enrich antigen‐specific high‐affinity B cells. The affinity of the membrane‐bound BCR in the flow chamber correlates well with the affinity of the matched antibodies measured in solution. These findings demonstrate that a microfluidic strategy can rapidly assess BCR‐antigen‐binding properties and identify antigen‐specific high‐affinity B cells. This strategy has the potential to both assess functional immune status from peripheral B cells and be a cost‐effective way of identifying individual B cells as antibody sources for a range of clinical applications.
Collapse
Affiliation(s)
- Ahmed M. Alhassan
- Department of Biomedical Engineering University of California Davis CA 95616 USA
| | - Venktesh S. Shirure
- Department of Biomedical Engineering University of California Davis CA 95616 USA
| | - Jean Luo
- Department of Pathology, Microbiology, and Immunology University of California Davis CA 95616 USA
| | - Bryan B. Nguyen
- Department of Biomedical Engineering University of California Davis CA 95616 USA
| | - Zachary A. Rollins
- Department of Biomedical Engineering University of California Davis CA 95616 USA
| | | | - Xiangdong Zhu
- Department of Physics and Astronomy University of California Davis CA 95616 USA
| | - Nicole Baumgarth
- Department of Pathology, Microbiology, and Immunology University of California Davis CA 95616 USA
- Department of Molecular Microbiology and Immunology Bloomberg School of Public Health and Department of Molecular and Comparative Pathobiology School of Medicine Johns Hopkins University Baltimore MD 21205 USA
| | - Steven C. George
- Department of Biomedical Engineering University of California Davis CA 95616 USA
| |
Collapse
|
12
|
Berndsen ZT, Akhtar M, Thapa M, Vickers T, Schmitz A, Torres JL, Baboo S, Kumar P, Khatoom N, Sheikh A, Hamrick M, Diedrich JK, Martinez-Bartolome S, Garrett PT, Yates JR, Turner JS, Laird RM, Poly F, Porter CK, Copps J, Ellebedy AH, Ward AB, Fleckenstein JM. Repeat modules and N-linked glycans define structure and antigenicity of a critical enterotoxigenic E. coli adhesin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.593125. [PMID: 38766097 PMCID: PMC11100705 DOI: 10.1101/2024.05.08.593125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Enterotoxigenic Escherichia coli (ETEC) cause hundreds of millions of cases of infectious diarrhea annually, predominantly in children from low-middle income regions. Notably, in children, as well as human volunteers challenged with ETEC, diarrheal severity is significantly increased severity in blood group A (bgA) individuals. EtpA, is a secreted glycoprotein adhesin that functions as a blood group A lectin to promote critical interactions between ETEC and blood group A glycans on intestinal epithelia for effective bacterial adhesion and toxin delivery. EtpA is highly immunogenic resulting in robust antibody responses following natural infection and experimental challenge of human volunteers with ETEC. To understand how EtpA directs ETEC-blood group A interactions and stimulates adaptive immunity, we mutated EtpA, mapped its glycosylation by mass-spectrometry (MS), isolated polyclonal (pAbs) and monoclonal antibodies (mAbs) from vaccinated mice and ETEC-infected human volunteers, and determined structures of antibody-EtpA complexes by cryo-electron microscopy. Both bgA and mAbs that inhibited EtpA-bgA interactions and ETEC adhesion, bound to the C-terminal repeat domain highlighting this region as crucial for ETEC pathogen-host interaction. MS analysis uncovered extensive and heterogeneous N-linked glycosylation of EtpA and cryo-EM structures revealed that mAbs directly engage these unique glycan containing epitopes. Finally, electron microscopy-based polyclonal epitope mapping revealed antibodies targeting numerous distinct epitopes on N and C-terminal domains, suggesting that EtpA vaccination generates responses against neutralizing and decoy regions of the molecule. Collectively, we anticipate that these data will inform our general understanding of pathogen-host glycan interactions and adaptive immunity relevant to rational vaccine subunit design.
Collapse
Affiliation(s)
- Zachary T Berndsen
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Marjahan Akhtar
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Mahima Thapa
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA. Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA and The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Tim Vickers
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Aaron Schmitz
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA. Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA and The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Jonathan L Torres
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Sabyasachi Baboo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Pardeep Kumar
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Nazia Khatoom
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Alaullah Sheikh
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Melissa Hamrick
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Jolene K Diedrich
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Patrick T Garrett
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - John R Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Jackson S Turner
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA. Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA and The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Renee M Laird
- Operationally Relevant Infections Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, USA
| | - Frédéric Poly
- Operationally Relevant Infections Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, USA
| | - Chad K Porter
- Naval Medical Research Command (NMRC), Silver Spring, Maryland, USA
- Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Jeffrey Copps
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Ali H Ellebedy
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA. Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA and The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - James M Fleckenstein
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
- Medicine Service, Infectious Diseases, Veterans Affairs Health Care System, Saint Louis Missouri, USA
| |
Collapse
|
13
|
Strazza V, Rossi M, Avati A, Tiseo G, Falcone M, Cusi MG, Menichetti F, Ricciardi-Castagnoli P, Tinti C, Pileri P. Rapid generation of human recombinant monoclonal antibodies from antibody-secreting cells using ferrofluid-based technology. Front Immunol 2024; 15:1341389. [PMID: 38698845 PMCID: PMC11064063 DOI: 10.3389/fimmu.2024.1341389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/06/2024] [Indexed: 05/05/2024] Open
Abstract
Monoclonal antibodies (mAbs) are one of the most important classes of biologics with high therapeutic and diagnostic value, but traditional methods for mAbs generation, such as hybridoma screening and phage display, have limitations, including low efficiency and loss of natural chain pairing. To overcome these challenges, novel single B cell antibody technologies have emerged, but they also have limitations such as in vitro differentiation of memory B cells and expensive cell sorters. In this study, we present a rapid and efficient workflow for obtaining human recombinant monoclonal antibodies directly from single antigen-specific antibody secreting cells (ASCs) in the peripheral blood of convalescent COVID-19 patients using ferrofluid technology. This process allows the identification and expression of recombinant antigen-specific mAbs in less than 10 days, using RT-PCR to generate linear Ig heavy and light chain gene expression cassettes, called "minigenes", for rapid expression of recombinant antibodies without cloning procedures. This approach has several advantages. First, it saves time and resources by eliminating the need for in vitro differentiation. It also allows individual antigen-specific ASCs to be screened for effector function prior to recombinant antibody cloning, enabling the selection of mAbs with desired characteristics and functional activity. In addition, the method allows comprehensive analysis of variable region repertoires in combination with functional assays to evaluate the specificity and function of the generated antigen-specific antibodies. Our approach, which rapidly generates recombinant monoclonal antibodies from single antigen-specific ASCs, could help to identify functional antibodies and deepen our understanding of antibody dynamics in the immune response through combined antibody repertoire sequence analysis and functional reactivity testing.
Collapse
Affiliation(s)
- Veronica Strazza
- Hyper Antibody Research & Development (HARD) -Lab, Toscana Life Sciences Foundation, Siena, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Marco Rossi
- Hyper Antibody Research & Development (HARD) -Lab, Toscana Life Sciences Foundation, Siena, Italy
| | - Andrea Avati
- Hyper Antibody Research & Development (HARD) -Lab, Toscana Life Sciences Foundation, Siena, Italy
| | - Giusy Tiseo
- Infectious Diseases Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliero Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Marco Falcone
- Infectious Diseases Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliero Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Maria Grazia Cusi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesco Menichetti
- Infectious Diseases Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliero Universitaria Pisana, University of Pisa, Pisa, Italy
| | | | - Cristina Tinti
- Hyper Antibody Research & Development (HARD) -Lab, Toscana Life Sciences Foundation, Siena, Italy
| | - Piero Pileri
- Hyper Antibody Research & Development (HARD) -Lab, Toscana Life Sciences Foundation, Siena, Italy
| |
Collapse
|
14
|
Lin Y, Wan Z, Liu B, Yao J, Li T, Yang F, Sui J, Zhao Y, Liu W, Zhou X, Wang J, Qi H. B cell-reactive triad of B cells, follicular helper and regulatory T cells at homeostasis. Cell Res 2024; 34:295-308. [PMID: 38326478 PMCID: PMC10978943 DOI: 10.1038/s41422-024-00929-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 01/08/2024] [Indexed: 02/09/2024] Open
Abstract
Autoreactive B cells are silenced through receptor editing, clonal deletion and anergy induction. Additional autoreactive B cells are ignorant because of physical segregation from their cognate autoantigen. Unexpectedly, we find that follicular B cell-derived autoantigen, including cell surface molecules such as FcγRIIB, is a class of homeostatic autoantigen that can induce spontaneous germinal centers (GCs) and B cell-reactive autoantibodies in non-autoimmune animals with intact T and B cell repertoires. These B cell-reactive B cells form GCs in a manner dependent on spontaneous follicular helper T (TFH) cells, which preferentially recognize B cell-derived autoantigen, and in a manner constrained by spontaneous follicular regulatory T (TFR) cells, which also carry specificities for B cell-derived autoantigen. B cell-reactive GC cells are continuously generated and, following immunization or infection, become intermixed with foreign antigen-induced GCs. Production of plasma cells and antibodies derived from B cell-reactive GC cells are markedly enhanced by viral infection, potentially increasing the chance for autoimmunity. Consequently, immune homeostasis in healthy animals not only involves classical tolerance of silencing and ignoring autoreactive B cells but also entails a reactive equilibrium attained by a spontaneous B cell-reactive triad of B cells, TFH cells and TFR cells.
Collapse
Affiliation(s)
- Yihan Lin
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Zurong Wan
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Weill Cornell Medical College, Cornell University, Ithaca, NY, USA
| | - Bo Liu
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Jiacheng Yao
- Changping Laboratory, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Tianqi Li
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Fang Yang
- National Institute of Biological Sciences, Beijing, China
| | - Jianhua Sui
- National Institute of Biological Sciences, Beijing, China
| | - Yongshan Zhao
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Wanli Liu
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Xuyu Zhou
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jianbin Wang
- Changping Laboratory, Beijing, China.
- School of Life Sciences, Tsinghua University, Beijing, China.
| | - Hai Qi
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
- Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing, China.
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China.
- Changping Laboratory, Beijing, China.
- New Cornerstone Science Laboratory, School of Medicine, Tsinghua University, Beijing, China.
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China.
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China.
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
15
|
Mallis RJ, Lee JJ, den Berg AV, Brazin KN, Viennet T, Zmuda J, Cross M, Radeva D, Rodriguez‐Mias R, Villén J, Gelev V, Reinherz EL, Arthanari H. Efficient and economic protein labeling for NMR in mammalian expression systems: Application to a preT-cell and T-cell receptor protein. Protein Sci 2024; 33:e4950. [PMID: 38511503 PMCID: PMC10955624 DOI: 10.1002/pro.4950] [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: 11/20/2023] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 03/22/2024]
Abstract
Protein nuclear magnetic resonance (NMR) spectroscopy relies on the ability to isotopically label polypeptides, which is achieved through heterologous expression in various host organisms. Most commonly, Escherichia coli is employed by leveraging isotopically substituted ammonium and glucose to uniformly label proteins with 15N and 13C, respectively. Moreover, E. coli can grow and express proteins in uniformly deuterium-substituted water (D2O), a strategy useful for experiments targeting high molecular weight proteins. Unfortunately, many proteins, particularly those requiring specific posttranslational modifications like disulfide bonding or glycosylation for proper folding and/or function, cannot be readily expressed in their functional forms using E. coli-based expression systems. One such class of proteins includes T-cell receptors and their related preT-cell receptors. In this study, we present an expression system for isotopic labeling of proteins using a nonadherent human embryonic kidney cell line, Expi293F, and a specially designed media. We demonstrate the application of this platform to the β subunit common to both receptors. In addition, we show that this expression system and media can be used to specifically label amino acids Phe, Ile, Val, and Leu in this system, utilizing an amino acid-specific labeling protocol that allows targeted incorporation at high efficiency without significant isotopic scrambling. We demonstrate that this system can also be used to express proteins with fluorinated amino acids. We were routinely able to obtain an NMR sample with a concentration of 200 μM from 30 mL of culture media, utilizing less than 20 mg of the labeled amino acids.
Collapse
Affiliation(s)
- Robert J. Mallis
- Laboratory of ImmunobiologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of DermatologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Jonathan J. Lee
- Laboratory of ImmunobiologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | | | - Kristine N. Brazin
- Laboratory of ImmunobiologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Thibault Viennet
- Department of Cancer BiologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical SchoolBostonMassachusettsUSA
| | | | | | - Denitsa Radeva
- Faculty of Chemistry and PharmacySofia UniversitySofiaBulgaria
| | | | - Judit Villén
- Department of Genome SciencesUniversity of WashingtonSeattleWashingtonUSA
| | - Vladimir Gelev
- Faculty of Chemistry and PharmacySofia UniversitySofiaBulgaria
| | - Ellis L. Reinherz
- Laboratory of ImmunobiologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Haribabu Arthanari
- Department of Cancer BiologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
16
|
Biswas S, Kuwata T, Yamauchi S, Okazaki K, Kaku Y, Hasan MZ, Morioka H, Matsushita S. Idiotopes of antibodies against HIV-1 CD4-induced epitope shared with those against microorganisms. Immunology 2024; 171:534-548. [PMID: 38102962 DOI: 10.1111/imm.13742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023] Open
Abstract
Induction of antibodies (Abs) against the conformational CD4-induced (CD4i) epitope is frequent in HIV-1 infection. However, the mechanism of development of anti-CD4i Abs is unclear. We used anti-idiotypic (aID) monoclonal Abs (mAbs) of anti-CD4i mAbs to isolate anti-CD4i mAbs from infected subjects and track the causative antigens. One anti-aID mAb sorted from infected subjects by aID mAbs had the characteristics of anti-CD4i Abs, including IGHV1-69 usage and ability to bind to HIV-1 Env enhanced by sCD4. Critical amino acid sequences for the binding of six anti-aID mAbs, with shared idiotope to anti-CD4i mAbs, were analysed by phage display. The identified amino acid sequences showed similarity to proteins from human microbiota and infectious agents. Peptides synthesized from Caudoviricetes sp and Vibrio vulnificus based on the identified sequences were reactive to most anti-aID and some anti-CD4i mAbs. These results suggest that anti-CD4i Abs may evolve from B cells primed by microorganisms.
Collapse
Affiliation(s)
- Shashwata Biswas
- Division of Clinical Retrovirology, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Takeo Kuwata
- Division of Clinical Retrovirology, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Soichiro Yamauchi
- Department of Analytical and Biophysical Chemistry, Faculty of Medical and Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kyo Okazaki
- Department of Analytical and Biophysical Chemistry, Faculty of Medical and Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yu Kaku
- Division of Clinical Retrovirology, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Md Zahid Hasan
- Division of Clinical Retrovirology, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Hiroshi Morioka
- Department of Analytical and Biophysical Chemistry, Faculty of Medical and Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shuzo Matsushita
- Division of Clinical Retrovirology, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
17
|
Li X, Zeng F, Yue R, Ma D, Meng Z, Li Q, Zhang Z, Zhang H, Liao Y, Liao Y, Jiang G, Zhao H, Yu L, Li D, Zhang Y, Liu L, Li Q. Heterologous Booster Immunization Based on Inactivated SARS-CoV-2 Vaccine Enhances Humoral Immunity and Promotes BCR Repertoire Development. Vaccines (Basel) 2024; 12:120. [PMID: 38400104 PMCID: PMC10891849 DOI: 10.3390/vaccines12020120] [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: 12/05/2023] [Revised: 01/13/2024] [Accepted: 01/17/2024] [Indexed: 02/25/2024] Open
Abstract
Recent studies have indicated that sequentially administering SARS-CoV-2 vaccines can result in increased antibody and cellular immune responses. In this study, we compared homologous and heterologous immunization strategies following two doses of inactivated vaccines in a mouse model. Our research demonstrates that heterologous sequential immunization resulted in more immune responses displayed in the lymph node germinal center, which induced a greater number of antibody-secreting cells (ASCs), resulting in enhanced humoral and cellular immune responses and increased cross-protection against five variant strains. In further single B-cell analysis, the above findings were supported by the presence of unique B-cell receptor (BCR) repertoires and diversity in CDR3 sequence profiles elicited by a heterologous booster immunization strategy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Longding Liu
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China (Y.Z.)
| | - Qihan Li
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China (Y.Z.)
| |
Collapse
|
18
|
Durgam SS, Khiew SH, Sayin I, Jain D, Yin D, Cavazzoni CB, Sage PT, King RG, Chong AS. MHC Tetramers Specifically Identify High- and Low-avidity Donor-specific B Cells in Transplantation Tolerance and Rejection. Transplantation 2023; 107:2526-2532. [PMID: 37493609 PMCID: PMC10811295 DOI: 10.1097/tp.0000000000004702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
BACKGROUND Although donor-specific antibody pre- and posttransplantation is routinely assessed, accurate quantification of memory alloreactive B cells that mediate recall antibody response remains challenging. Major histocompatibility complex (MHC) tetramers have been used to identify alloreactive B cells in mice and humans, but the specificity of this approach has not been rigorously assessed. METHODS B-cell receptors from MHC tetramer-binding single B cells were expressed as mouse recombinant immunoglobulin G1 (rIgG1) monoclonal antibodies, and the specificity was assessed with a multiplex bead assay. Relative binding avidity of rIgG1 was measured by modified dilution series technique and surface plasmon resonance. Additionally, immunoglobulin heavy chain variable regions of 50 individual B-cell receptors were sequenced to analyze the rate of somatic hypermutation. RESULTS The multiplex bead assay confirmed that expressed rIgG1 monoclonal antibodies were preferentially bound to bait MHC class II I-E d over control I-A d and I-A b tetramers. Furthermore, the dissociation constant 50 binding avidities of the rIgG1 ranged from 10 mM to 7 nM. The majority of tetramer-binding B cells were low avidity, and ~12.8% to 15.2% from naive and tolerant mice and 30.9% from acute rejecting mice were higher avidity (dissociation constant 50 <1 mM). CONCLUSIONS Collectively, these studies demonstrate that donor MHC tetramers, under stringent binding conditions with decoy self-MHC tetramers, can specifically identify a broad repertoire of donor-specific B cells under conditions of rejection and tolerance.
Collapse
Affiliation(s)
| | | | - Ismail Sayin
- Department of Surgery, The University of Chicago, Chicago,
IL, USA
| | - Dharmendra Jain
- Department of Surgery, The University of Chicago, Chicago,
IL, USA
| | - Dengping Yin
- Department of Surgery, The University of Chicago, Chicago,
IL, USA
| | - Cecilia B. Cavazzoni
- Transplantation Research Center, Renal Division, Brigham
and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter T. Sage
- Transplantation Research Center, Renal Division, Brigham
and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - R. Glenn King
- Department of Microbiology, University of Alabama at
Birmingham, Birmingham, AL, 35294, USA
| | - Anita S Chong
- Department of Surgery, The University of Chicago, Chicago,
IL, USA
| |
Collapse
|
19
|
Tan K, Chen J, Kaku Y, Wang Y, Donius L, Khan RA, Li X, Richter H, Seaman MS, Walz T, Hwang W, Reinherz EL, Kim M. Inadequate structural constraint on Fab approach rather than paratope elicitation limits HIV-1 MPER vaccine utility. Nat Commun 2023; 14:7218. [PMID: 37940661 PMCID: PMC10632514 DOI: 10.1038/s41467-023-42097-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/25/2023] [Indexed: 11/10/2023] Open
Abstract
Broadly neutralizing antibodies (bnAbs) against HIV-1 target conserved envelope (Env) epitopes to block viral replication. Here, using structural analyses, we provide evidence to explain why a vaccine targeting the membrane-proximal external region (MPER) of HIV-1 elicits antibodies with human bnAb-like paratopes paradoxically unable to bind HIV-1. Unlike in natural infection, vaccination with MPER/liposomes lacks a necessary structure-based constraint to select for antibodies with an adequate approach angle. Consequently, the resulting Abs cannot physically access the MPER crawlspace on the virion surface. By studying naturally arising Abs, we further reveal that flexibility of the human IgG3 hinge mitigates the epitope inaccessibility and additionally facilitates Env spike protein crosslinking. Our results suggest that generation of IgG3 subtype class-switched B cells is a strategy for anti-MPER bnAb induction. Moreover, the findings illustrate the need to incorporate topological features of the target epitope in immunogen design.
Collapse
Affiliation(s)
- Kemin Tan
- Structural Biology Center, X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Lemont, IL, USA
| | - Junjian Chen
- Laboratory of Immunobiology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Laboratory of Immunology, Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yu Kaku
- Laboratory of Immunobiology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Yi Wang
- Laboratory of Immunobiology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- NeoCura Bio-Medical Technology Co., Ltd., Beijing, China
| | - Luke Donius
- Laboratory of Immunobiology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- AbbVie Bioresearch Center, AbbVie Inc., Worcester, MA, USA
| | - Rafiq Ahmad Khan
- Laboratory of Immunobiology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Xiaolong Li
- Laboratory of Immunobiology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Hannah Richter
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Michael S Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Thomas Walz
- Laboratory of Molecular Electron Microscopy, The Rockefeller University, New York, NY, USA
| | - Wonmuk Hwang
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
- Department of Materials Science & Engineering, Texas A&M University, College Station, TX, USA
- Department of Physics & Astronomy, Texas A&M University, College Station, TX, USA
| | - Ellis L Reinherz
- Laboratory of Immunobiology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Mikyung Kim
- Laboratory of Immunobiology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Dermatology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
New JS, Dizon BL, King RG, Greenspan NS, Kearney JF. B-1 B Cell-Derived Natural Antibodies against N-Acetyl-d-Glucosamine Suppress Autoimmune Diabetes Pathogenesis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1320-1331. [PMID: 37747293 PMCID: PMC10592000 DOI: 10.4049/jimmunol.2300264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023]
Abstract
Environmental factors and host microbiota strongly influence type 1 diabetes (T1D) progression. We report that neonatal immunization with group A Streptococcus suppresses T1D development in NOD mice by promoting clonal expansion of N-acetyl-d-glucosamine (GlcNAc)-specific B-1 B cells that recognize pancreatic β cell-derived Ags bearing GlcNAc-containing posttranslational modifications. Early exposure to Lancefield group A cell-wall carbohydrate Ags increased production of GlcNAc-reactive serum Abs and enhanced localization of innate-like GlcNAc-specific B cells to pancreatic tissue during T1D pathogenesis. We show that B-1 B cell-derived GlcNAc-specific IgM engages apoptosis-associated β cell Ags, thereby suppressing diabetogenic T cell activation. Likewise, adoptively transferring GlcNAc-reactive B-1 B cells significantly delayed T1D development in naive recipients. Collectively, these data underscore potentially protective involvement of innate-like B cells and natural Abs in T1D progression. These findings suggest that previously reported associations of reduced T1D risk after GAS infection are B cell dependent and demonstrate the potential for targeting the natural Ab repertoire in considering therapeutic strategies for T1D.
Collapse
Affiliation(s)
- J. Stewart New
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Brian L.P. Dizon
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - R. Glenn King
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Neil. S. Greenspan
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - John F. Kearney
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
21
|
Cousu C, Mulot E, De Smet A, Formichetti S, Lecoeuche D, Ren J, Muegge K, Boulard M, Weill JC, Reynaud CA, Storck S. Germinal center output is sustained by HELLS-dependent DNA-methylation-maintenance in B cells. Nat Commun 2023; 14:5695. [PMID: 37709749 PMCID: PMC10502085 DOI: 10.1038/s41467-023-41317-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 08/29/2023] [Indexed: 09/16/2023] Open
Abstract
HELLS/LSH (Helicase, Lymphoid Specific) is a SNF2-like chromatin remodelling protein involved in DNA methylation. Its loss-of-function in humans causes humoral immunodeficiency, called ICF4 syndrome (Immunodeficiency, Centromeric Instability, Facial anomalies). Here we show by our newly generated B-cell-specific Hells conditional knockout mouse model that HELLS plays a pivotal role in T-dependent B-cell responses. HELLS deficiency induces accelerated decay of germinal center (GC) B cells and impairs the generation of high affinity memory B cells and circulating antibodies. Mutant GC B cells undergo dramatic DNA hypomethylation and massive de-repression of evolutionary recent retrotransposons, which surprisingly does not directly affect their survival. Instead, they prematurely upregulate either memory B cell markers or the transcription factor ATF4, which is driving an mTORC1-dependent metabolic program typical of plasma cells. Treatment of wild type mice with a DNMT1-specific inhibitor phenocopies the accelerated kinetics, thus pointing towards DNA-methylation maintenance by HELLS being a crucial mechanism to fine-tune the GC transcriptional program and enable long-lasting humoral immunity.
Collapse
Affiliation(s)
- Clara Cousu
- Université Paris Cité, CNRS UMR 8253, INSERM U1151, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Eléonore Mulot
- Université Paris Cité, CNRS UMR 8253, INSERM U1151, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Annie De Smet
- Université Paris Cité, CNRS UMR 8253, INSERM U1151, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Sara Formichetti
- Epigenetics and Neurobiology Unit, European Molecular Biology Laboratory (EMBL), 00015, Monterotondo, Italy
- Joint PhD degree program, European Molecular Biology Laboratory and Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Damiana Lecoeuche
- Université Paris Cité, CNRS UMR 8253, INSERM U1151, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Jianke Ren
- Epigenetics Section, Frederick National Laboratory for Cancer Research in the Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
- NHC Key Lab of Reproduction Regulation,Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, 200237, China
| | - Kathrin Muegge
- Epigenetics Section, Frederick National Laboratory for Cancer Research in the Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA
| | - Matthieu Boulard
- Epigenetics and Neurobiology Unit, European Molecular Biology Laboratory (EMBL), 00015, Monterotondo, Italy
| | - Jean-Claude Weill
- Université Paris Cité, CNRS UMR 8253, INSERM U1151, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Claude-Agnès Reynaud
- Université Paris Cité, CNRS UMR 8253, INSERM U1151, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Sébastien Storck
- Université Paris Cité, CNRS UMR 8253, INSERM U1151, Institut Necker Enfants Malades, F-75015, Paris, France.
| |
Collapse
|
22
|
Alhassan AM, Shirure VS, Luo J, Nguyen BB, Rollins ZA, Shergill BS, Zhu X, Baumgarth N, George SC. A microfluidic strategy to capture antigen-specific high affinity B cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.12.548739. [PMID: 37503139 PMCID: PMC10369944 DOI: 10.1101/2023.07.12.548739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Assessing B cell affinity to pathogen-specific antigens prior to or following exposure could facilitate the assessment of immune status. Current standard tools to assess antigen-specific B cell responses focus on equilibrium binding of the secreted antibody in serum. These methods are costly, time-consuming, and assess antibody affinity under zero-force. Recent findings indicate that force may influence BCR-antigen binding interactions and thus immune status. Here, we designed a simple laminar flow microfluidic chamber in which the antigen (hemagglutinin of influenza A) is bound to the chamber surface to assess antigen-specific BCR binding affinity of five hemagglutinin-specific hybridomas under 65- to 650-pN force range. Our results demonstrate that both increasing shear force and bound lifetime can be used to enrich antigen-specific high affinity B cells. The affinity of the membrane-bound BCR in the flow chamber correlates well with the affinity of the matched antibodies measured in solution. These findings demonstrate that a microfluidic strategy can rapidly assess BCR-antigen binding properties and identify antigen-specific high affinity B cells. This strategy has the potential to both assess functional immune status from peripheral B cells and be a cost-effective way of identifying individual B cells as antibody sources for a range of clinical applications.
Collapse
Affiliation(s)
- Ahmed M. Alhassan
- Department of Biomedical Engineering, University of California, Davis
| | | | - Jean Luo
- Department of Pathology, Microbiology, and Immunology, University of California, Davis
| | - Bryan B. Nguyen
- Department of Biomedical Engineering, University of California, Davis
| | | | | | - Xiangdong Zhu
- Department of Physics and Astronomy, University of California, Davis
| | - Nicole Baumgarth
- Department of Pathology, Microbiology, and Immunology, University of California, Davis
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health and Department of Molecular and Comparative Pathobiology, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Steven C. George
- Department of Biomedical Engineering, University of California, Davis
| |
Collapse
|
23
|
Tan K, Chen J, Kaku Y, Wang Y, Donius L, Khan RA, Li X, Richter H, Seaman MS, Walz T, Hwang W, Reinherz EL, Kim M. Inadequate structural constraint on Fab approach rather than paratope elicitation limits HIV-1 MPER vaccine utility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.27.546734. [PMID: 37425731 PMCID: PMC10327024 DOI: 10.1101/2023.06.27.546734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Broadly neutralizing antibodies (bnAbs) against HIV-1 target conserved epitopes, thereby inhibiting viral entry. Yet surprisingly, those recognizing linear epitopes in the HIV-1 gp41 membrane proximal external region (MPER) are elicited neither by peptide nor protein scaffold vaccines. Here, we observe that while Abs generated by MPER/liposome vaccines may exhibit human bnAb-like paratopes, B-cell programming without constraints imposed by the gp160 ectodomain selects Abs unable to access the MPER within its native "crawlspace". During natural infection, the flexible hinge of IgG3 partially mitigates steric occlusion of less pliable IgG1 subclass Abs with identical MPER specificity, until affinity maturation refines entry mechanisms. The IgG3 subclass maintains B-cell competitiveness, exploiting bivalent ligation resulting from greater intramolecular Fab arm length, offsetting weak antibody affinity. These findings suggest future immunization strategies.
Collapse
|
24
|
Fujisawa M, Adachi Y, Onodera T, Shiwa-Sudo N, Iwata-Yoshikawa N, Nagata N, Suzuki T, Takeoka S, Takahashi Y. High-throughput isolation of SARS-CoV-2 nucleocapsid antibodies for improved antigen detection. Biochem Biophys Res Commun 2023; 673:114-120. [PMID: 37379800 PMCID: PMC10279465 DOI: 10.1016/j.bbrc.2023.06.067] [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: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
SARS-CoV-2 nucleocapsid protein (NP) is the main target for COVID-19-diagnostic PCR and antigen rapid diagnostic tests (Ag-RDTs). Ag-RDTs are more convenient than PCR tests for point-of-care testing or self-testing to identify the SARS-CoV-2 antigen. The sensitivity and specificity of this method depends mainly on the affinity and specificity of NP-binding antibodies; therefore, antigen-antibody binding is key elements for the Ag-RDTs. Here, we applied the high-throughput antibody isolation platform that has been utilized to isolate therapeutic antibodies against rare epitopes. Two NP antibodies were identified to recognize non-overlapping epitopes with high affinity. One antibody specifically binds to SARS-CoV-2 NP, and the other rapidly and tightly binds to SARS-CoV-2 NP with cross-reactivity to SARS-CoV NP. Furthermore, these antibodies were compatible with a sandwich enzyme-linked immunosorbent assay that exhibited enhanced sensitivity for NP detection compared to the previously isolated NP antibodies. Thus, the NP antibody pair is applicable to more sensitive and specific Ag-RDTs, highlighting the utility of a high-throughput antibody isolation platform for diagnostics development.
Collapse
Affiliation(s)
- Mizuki Fujisawa
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan; Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Yu Adachi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Taishi Onodera
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Nozomi Shiwa-Sudo
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama-shi, Tokyo, 208-0011, Japan
| | - Naoko Iwata-Yoshikawa
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama-shi, Tokyo, 208-0011, Japan
| | - Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama-shi, Tokyo, 208-0011, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashi-murayama-shi, Tokyo, 208-0011, Japan
| | - Shinji Takeoka
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo, 162-8480, Japan; Research Institute for Science and Engineering, Waseda University, 3-4-1, Ohkubo, Shinjuku-ku, Tokyo, 169-8555, Japan
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan.
| |
Collapse
|
25
|
Caniels TG, Medina-Ramírez M, Zhang J, Sarkar A, Kumar S, LaBranche A, Derking R, Allen JD, Snitselaar JL, Capella-Pujol J, Sánchez IDM, Yasmeen A, Diaz M, Aldon Y, Bijl TPL, Venkatayogi S, Martin Beem JS, Newman A, Jiang C, Lee WH, Pater M, Burger JA, van Breemen MJ, de Taeye SW, Rantalainen K, LaBranche C, Saunders KO, Montefiori D, Ozorowski G, Ward AB, Crispin M, Moore JP, Klasse PJ, Haynes BF, Wilson IA, Wiehe K, Verkoczy L, Sanders RW. Germline-targeting HIV-1 Env vaccination induces VRC01-class antibodies with rare insertions. Cell Rep Med 2023; 4:101003. [PMID: 37044090 PMCID: PMC10140475 DOI: 10.1016/j.xcrm.2023.101003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/23/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023]
Abstract
Targeting germline (gl-) precursors of broadly neutralizing antibodies (bNAbs) is acknowledged as an important strategy for HIV-1 vaccines. The VRC01-class of bNAbs is attractive because of its distinct genetic signature. However, VRC01-class bNAbs often require extensive somatic hypermutation, including rare insertions and deletions. We describe a BG505 SOSIP trimer, termed GT1.2, to optimize binding to gl-CH31, the unmutated common precursor of the CH30-34 bNAb lineage that acquired a large CDRH1 insertion. The GT1.2 trimer activates gl-CH31 naive B cells in knock-in mice, and B cell responses could be matured by selected boosting immunogens to generate cross-reactive Ab responses. Next-generation B cell sequencing reveals selection for VRC01-class mutations, including insertions in CDRH1 and FWR3 at positions identical to VRC01-class bNAbs, as well as CDRL1 deletions and/or glycine substitutions to accommodate the N276 glycan. These results provide proof of concept for vaccine-induced affinity maturation of B cell lineages that require rare insertions and deletions.
Collapse
Affiliation(s)
- Tom G Caniels
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - Max Medina-Ramírez
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - Jinsong Zhang
- Applied Biomedical Science Institute, San Diego, CA, USA
| | - Anita Sarkar
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Sonu Kumar
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Alex LaBranche
- Applied Biomedical Science Institute, San Diego, CA, USA
| | - Ronald Derking
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - Joel D Allen
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Jonne L Snitselaar
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - Joan Capella-Pujol
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - Iván Del Moral Sánchez
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - Anila Yasmeen
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA
| | - Marilyn Diaz
- Applied Biomedical Science Institute, San Diego, CA, USA
| | - Yoann Aldon
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - Tom P L Bijl
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | | | | | - Amanda Newman
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Chuancang Jiang
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA
| | - Wen-Hsin Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Maarten Pater
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - Judith A Burger
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - Mariëlle J van Breemen
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - Steven W de Taeye
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands
| | - Kimmo Rantalainen
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Celia LaBranche
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Kevin O Saunders
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - David Montefiori
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA; Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Gabriel Ozorowski
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - John P Moore
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA
| | - Per Johan Klasse
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA
| | - Barton F Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, 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
| | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | | | - Rogier W Sanders
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, the Netherlands; Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY, USA.
| |
Collapse
|
26
|
Nihei Y, Haniuda K, Higashiyama M, Asami S, Iwasaki H, Fukao Y, Nakayama M, Suzuki H, Kikkawa M, Kazuno S, Miura Y, Suzuki Y, Kitamura D. Identification of IgA autoantibodies targeting mesangial cells redefines the pathogenesis of IgA nephropathy. SCIENCE ADVANCES 2023; 9:eadd6734. [PMID: 36947618 PMCID: PMC10032602 DOI: 10.1126/sciadv.add6734] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 02/21/2023] [Indexed: 06/18/2023]
Abstract
Immunoglobulin A (IgA) nephropathy (IgAN) is the most common type of primary glomerulonephritis, often progressing to renal failure. IgAN is triggered by IgA deposition in the glomerular mesangium by an undefined mechanism. Here, we show that grouped ddY (gddY) mice, a spontaneous IgAN model, produce serum IgA against mesangial antigens, including βII-spectrin. Most patients with IgAN also have serum anti-βII-spectrin IgA. As in patients with IgAN, IgA+ plasmablasts accumulate in the kidneys of gddY mice. IgA antibodies cloned from the plasmablasts carry substantial V-region mutations and bind to βII-spectrin and the surface of mesangial cells. These IgAs recognize transfected and endogenous βII-spectrin exposed on the surface of embryonic kidney-derived cells. Last, we demonstrate that the cloned IgA can bind selectively to glomerular mesangial regions in situ. The identification of IgA autoantibody and its antigen in IgAN provides key insights into disease onset and redefines IgAN as a tissue-specific autoimmune disease.
Collapse
Affiliation(s)
- Yoshihito Nihei
- Department of Nephrology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
- Division of Cancer Cell Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo 278-0022, Japan
| | - Kei Haniuda
- Division of Cancer Cell Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo 278-0022, Japan
| | - Mizuki Higashiyama
- Division of Cancer Cell Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo 278-0022, Japan
| | - Shohei Asami
- Division of Cancer Cell Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo 278-0022, Japan
| | - Hiroyuki Iwasaki
- Department of Nephrology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
- Division of Cancer Cell Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo 278-0022, Japan
| | - Yusuke Fukao
- Department of Nephrology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Maiko Nakayama
- Department of Nephrology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Hitoshi Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Mika Kikkawa
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Saiko Kazuno
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yoshiki Miura
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Daisuke Kitamura
- Division of Cancer Cell Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo 278-0022, Japan
| |
Collapse
|
27
|
Chen ST, Oliveira TY, Gazumyan A, Cipolla M, Nussenzweig MC. B cell receptor signaling in germinal centers prolongs survival and primes B cells for selection. Immunity 2023; 56:547-561.e7. [PMID: 36882061 PMCID: PMC10424567 DOI: 10.1016/j.immuni.2023.02.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 10/28/2022] [Accepted: 02/08/2023] [Indexed: 03/08/2023]
Abstract
Germinal centers (GCs) are sites of B cell clonal expansion, diversification, and antibody affinity selection. This process is limited and directed by T follicular helper cells that provide helper signals to B cells that endocytose, process, and present cognate antigens in proportion to their B cell receptor (BCR) affinity. Under this model, the BCR functions as an endocytic receptor for antigen capture. How signaling through the BCR contributes to selection is not well understood. To investigate the role of BCR signaling in GC selection, we developed a tracker for antigen binding and presentation and a Bruton's tyrosine kinase drug-resistant-mutant mouse model. We showed that BCR signaling per se is necessary for the survival and priming of light zone B cells to receive T cell help. Our findings provide insight into how high-affinity antibodies are selected within GCs and are fundamental to our understanding of adaptive immunity and vaccine development.
Collapse
Affiliation(s)
- Spencer T Chen
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA.
| | - Thiago Y Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Anna Gazumyan
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Melissa Cipolla
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute (HHMI), The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
28
|
Nakao H, Yamaguchi T, Kawabata K, Higashi K, Nonaka M, Tuiji M, Nagai Y, Toyoda H, Yamaguchi Y, Kawasaki N, Kawasaki T. Characterization of novel antibodies that recognize sialylated keratan sulfate and lacto-N-fucopentaose I on human induced pluripotent cells: comparison with existing antibodies. Glycobiology 2023; 33:150-164. [PMID: 36373215 DOI: 10.1093/glycob/cwac074] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 10/24/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
This report describes the isolation and characterization of two new antibodies, R-6C (IgM) and R-13E (IgM), which were generated in C57BL/6 mice (Mus musculus) using the Tic (JCRB1331) human induced pluripotent cell (hiPSC) line as an antigen, and their comparisons with two existing antibodies, R-10G (IgG1) and R-17F (IgG1). Their epitopes were studied by western blotting after various glycosidase digestions, binding analyses using enzyme-linked immunosorbent assays (ELISAs) and microarrays with various synthetic oligosaccharides. The minimum epitope structures identified were: Siaα2-3Galβ1-3GlcNAc(6S)β1-3Galβ1-4GlcNAc(6S)β1 (R-6C), Fucα1-2Galβ1-3GlcNAcβ1-3Galβ1 (R-13E), Galβ1-4GlcNAc(6S)β1-3Galβ1-4GlcNAc(6S)β1 (R-10G), and Fucα1-2Galβ1-3GlcNAβ1-3Galβ1-4Glc (lacto-N-fucopentaose I) (R-17F). Most glycoprotein epitopes are expressed as O-glycans. The common feature of these epitopes is the presence of an N-acetyllactosamine type 1 structure (Galβ1-3GlcNAc) at their nonreducing termini, followed by a type 2 structure (Galβ1-4GlcNAc); this arrangement comprises a type 1-type 2 motif. This motif is also shared by TRA-1-60, a traditional onco-fetal antigen. In contrast, the R-10G epitope has a type 2-type 2 motif. Among these antibodies, R-17F and R-13E exhibit cytotoxic activity toward hiPSCs. R-17F and R-13E exhibit extremely high similarity in the amino acid sequences in their complementarity-determining regions (CDRs), which is consistent with their highly similar glycan recognition. These antibodies are excellent tools for investigating the biological functions of glycoconjugates in hiPSCs/hESCs; they could be useful for the selection, isolation and selective killing of such undifferentiated pluripotent stem cells.
Collapse
Affiliation(s)
- Hiromi Nakao
- Glycobiotechnology Laboratory, Research Organization of Science and Technology, Ritsumeikan University, Noji-Higashi 1-1-1, Kusatsu, Shiga 525-8577, Japan
| | - Tomoko Yamaguchi
- Laboratory of Cell Model for Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Saito-Asagi 7-6-8, Ibaraki, Osaka 567-0085, Japan
| | - Kenji Kawabata
- Laboratory of Cell Model for Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Saito-Asagi 7-6-8, Ibaraki, Osaka 567-0085, Japan
| | - Katsuaki Higashi
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Shogoin-Kawaharacho 53, Sakyo-ku, Kyoto, Kyoto 606-8507, Japan
| | - Motohiro Nonaka
- Department of Biological Chemistry, Human Health Sciences, Graduate School of Medicine, Kyoto University, Shogoin-Kawaharacho 53, Sakyo-ku, Kyoto, Kyoto 606-8507, Japan
| | - Makoto Tuiji
- Department of Microbiology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Ebara 2-4-41, Shinagawa-ku, Tokyo 142-8501, Japan
| | - Yuko Nagai
- Laboratory of Bio-analytical Chemistry, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga 525-8577, Japan
| | - Hidenao Toyoda
- Laboratory of Bio-analytical Chemistry, College of Pharmaceutical Sciences, Ritsumeikan University, Shiga 525-8577, Japan
| | - Yoshiki Yamaguchi
- Division of Structural Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Komatsushima 4-4-1, Aobaku, Sendai, Miyagi 981-8558, Japan
| | - Nobuko Kawasaki
- Glycobiotechnology Laboratory, Research Organization of Science and Technology, Ritsumeikan University, Noji-Higashi 1-1-1, Kusatsu, Shiga 525-8577, Japan
| | - Toshisuke Kawasaki
- Glycobiotechnology Laboratory, Research Organization of Science and Technology, Ritsumeikan University, Noji-Higashi 1-1-1, Kusatsu, Shiga 525-8577, Japan.,Laboratory of Cell Model for Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Saito-Asagi 7-6-8, Ibaraki, Osaka 567-0085, Japan
| |
Collapse
|
29
|
McKenzie CI, Varese N, Aui PM, Reinwald S, Wines BD, Hogarth PM, Thien F, Hew M, Rolland JM, O'Hehir RE, van Zelm MC. RNA sequencing of single allergen-specific memory B cells after grass pollen immunotherapy: Two unique cell fates and CD29 as a biomarker for treatment effect. Allergy 2023; 78:822-835. [PMID: 36153670 PMCID: PMC10952829 DOI: 10.1111/all.15529] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Sublingual immunotherapy (SLIT) for grass pollen allergy can modify the natural history of allergic rhinitis and is associated with increased allergen-specific IgG4 . IgG4 competitively inhibits functional IgE on the surface of effector cells, such as mast cells and basophils, from binding to allergens. To further understand the important role memory B-cell (Bmem) responses play in mediating the beneficial effects of SLIT, we assessed changes in allergen-specific Bmem subsets induced by SLIT for grass pollen allergy. METHODS Blood samples were collected twice outside the pollen season from twenty-seven patients with sensitization to ryegrass pollen (RGP; Lolium perenne) and seasonal rhinoconjunctivitis. Thirteen received 4-month pre-seasonal SLIT for grass pollen allergy, and 14 received standard pharmacotherapy only. Single-cell RNA sequencing was performed on FACS-purified Lol p 1-specific Bmem before and after SLIT from four patients, and significant genes were validated by flow cytometry on the total cohort. RESULTS Four months of SLIT increased RGP-specific IgE and IgG4 in serum and induced two Lol p 1-specific Bmem subsets with unique transcriptional profiles. Both subsets had upregulated expression of beta 1 integrin ITGB1 (CD29), whereas IGHE (IgE), IGHG4 (IgG4 ), FCER2 (CD23), and IL13RA1 were upregulated in one subset. There was an increase in the proportion of Lol p 1+ Bmem expressing surface IgG4 , CD23, and CD29 after SLIT. CONCLUSIONS A clinically successful 4 months course of SLIT for grass pollen allergy induces two transcriptionally unique Bmem fates. Associated changes in surface-expressed proteins on these Bmem subsets can be used as early biomarkers for treatment effects.
Collapse
Affiliation(s)
- Craig I. McKenzie
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Nirupama Varese
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Pei Mun Aui
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Simone Reinwald
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Bruce D. Wines
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
- Department of PathologyThe University of MelbourneParkvilleVictoriaAustralia
| | - P. Mark Hogarth
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
- Department of PathologyThe University of MelbourneParkvilleVictoriaAustralia
| | - Francis Thien
- Respiratory Medicine, Eastern HealthBox Hill and Monash UniversityMelbourneVictoriaAustralia
| | - Mark Hew
- School of Public Health and Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
- Allergy, Asthma and Clinical ImmunologyAlfred HealthMelbourneVictoriaAustralia
| | - Jennifer M. Rolland
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Robyn E. O'Hehir
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Allergy, Asthma and Clinical ImmunologyAlfred HealthMelbourneVictoriaAustralia
| | - Menno C. van Zelm
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Allergy, Asthma and Clinical ImmunologyAlfred HealthMelbourneVictoriaAustralia
| |
Collapse
|
30
|
Kuraoka M, Aschner CB, Windsor IW, Mahant AM, Garforth SJ, Kong SL, Achkar JM, Almo SC, Kelsoe G, Herold BC. A non-neutralizing glycoprotein B monoclonal antibody protects against herpes simplex virus disease in mice. J Clin Invest 2023; 133:e161968. [PMID: 36454639 PMCID: PMC9888390 DOI: 10.1172/jci161968] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
There is an unmet need for monoclonal antibodies (mAbs) for prevention or as adjunctive treatment of herpes simplex virus (HSV) disease. Most vaccine and mAb efforts focus on neutralizing antibodies, but for HSV this strategy has proven ineffective. Preclinical studies with a candidate HSV vaccine strain, ΔgD-2, demonstrated that non-neutralizing antibodies that activate Fcγ receptors (FcγRs) to mediate antibody-dependent cellular cytotoxicity (ADCC) provide active and passive protection against HSV-1 and HSV-2. We hypothesized that this vaccine provides a tool to identify and characterize protective mAbs. We isolated HSV-specific mAbs from germinal center and memory B cells and bone marrow plasmacytes of ΔgD-2-vaccinated mice and evaluated these mAbs for binding, neutralizing, and FcγR-activating activity and for protective efficacy in mice. The most potent protective mAb, BMPC-23, was not neutralizing but activated murine FcγRIV, a biomarker of ADCC. The cryo-electron microscopic structure of the Fab-glycoprotein B (gB) assembly identified domain IV of gB as the epitope. A single dose of BMPC-23 administered 24 hours before or after viral challenge provided significant protection when configured as mouse IgG2c and protected mice expressing human FcγRIII when engineered as a human IgG1. These results highlight the importance of FcR-activating antibodies in protecting against HSV.
Collapse
Affiliation(s)
- Masayuki Kuraoka
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Clare Burn Aschner
- Department of Microbiology-Immunology, Albert Einstein College of Medicine, New York, New York, USA
| | - Ian W. Windsor
- Department of Laboratory of Molecular Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Aakash Mahant Mahant
- Department of Microbiology-Immunology, Albert Einstein College of Medicine, New York, New York, USA
| | | | - Susan Luozheng Kong
- Department of Laboratory of Molecular Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jacqueline M. Achkar
- Department of Microbiology-Immunology, Albert Einstein College of Medicine, New York, New York, USA
- Department of Medicine, Albert Einstein College of Medicine, New York, New York, USA
| | | | - Garnett Kelsoe
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Surgery and
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Betsy C. Herold
- Department of Microbiology-Immunology, Albert Einstein College of Medicine, New York, New York, USA
- Department of Pediatrics Albert Einstein College of Medicine, New York, New York, USA
| |
Collapse
|
31
|
Leonaviciene G, Mazutis L. RNA cytometry of single-cells using semi-permeable microcapsules. Nucleic Acids Res 2023; 51:e2. [PMID: 36268865 PMCID: PMC9841424 DOI: 10.1093/nar/gkac918] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/23/2022] [Accepted: 10/07/2022] [Indexed: 01/29/2023] Open
Abstract
Analytical tools for gene expression profiling of individual cells are critical for studying complex biological systems. However, the techniques enabling rapid measurements of gene expression on thousands of single-cells are lacking. Here, we report a high-throughput RNA cytometry for digital profiling of single-cells isolated in liquid droplets enveloped by a thin semi-permeable membrane (microcapsules). Due to the selective permeability of the membrane, the desirable enzymes and reagents can be loaded, or replaced, in the microcapsule at any given step by simply changing the reaction buffer in which the microcapsules are dispersed. Therefore, complex molecular biology workflows can be readily adapted to conduct nucleic acid analysis on encapsulated mammalian cells, or other biological species. The microcapsules support sequential multi-step enzymatic reactions and remain intact under different biochemical conditions, freezing, thawing, and thermocycling. Combining microcapsules with conventional FACS provides a high-throughput approach for conducting RNA cytometry of individual cells based on their digital gene expression signature.
Collapse
Affiliation(s)
- Greta Leonaviciene
- Institute of Biotechnology, Life Sciences Centre, Vilnius University, 7 Sauletekio av., Vilnius, LT-10257, Lithuania
| | - Linas Mazutis
- Institute of Biotechnology, Life Sciences Centre, Vilnius University, 7 Sauletekio av., Vilnius, LT-10257, Lithuania
| |
Collapse
|
32
|
Caradonna TM, Ronsard L, Yousif AS, Windsor IW, Hecht R, Bracamonte-Moreno T, Roffler AA, Maron MJ, Maurer DP, Feldman J, Marchiori E, Barnes RM, Rohrer D, Lonberg N, Oguin TH, Sempowski GD, Kepler TB, Kuraoka M, Lingwood D, Schmidt AG. An epitope-enriched immunogen expands responses to a conserved viral site. Cell Rep 2022; 41:111628. [PMID: 36351401 PMCID: PMC9883670 DOI: 10.1016/j.celrep.2022.111628] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 08/22/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022] Open
Abstract
Pathogens evade host humoral responses by accumulating mutations in surface antigens. While variable, there are conserved regions that cannot mutate without compromising fitness. Antibodies targeting these conserved epitopes are often broadly protective but remain minor components of the repertoire. Rational immunogen design leverages a structural understanding of viral antigens to modulate humoral responses to favor these responses. Here, we report an epitope-enriched immunogen presenting a higher copy number of the influenza hemagglutinin (HA) receptor-binding site (RBS) epitope relative to other B cell epitopes. Immunization in a partially humanized murine model imprinted with an H1 influenza shows H1-specific serum and >99% H1-specific B cells being RBS-directed. Single B cell analyses show a genetically restricted response that structural analysis defines as RBS-directed antibodies engaging the RBS with germline-encoded contacts. These data show how epitope enrichment expands B cell responses toward conserved epitopes and advances immunogen design approaches for next-generation viral vaccines.
Collapse
Affiliation(s)
| | - Larance Ronsard
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Ashraf S Yousif
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | | | - Rachel Hecht
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | | | - Anne A Roffler
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Max J Maron
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Daniel P Maurer
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Jared Feldman
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Elisa Marchiori
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Ralston M Barnes
- Bristol-Myers Squibb, 700 Bay Road, Redwood City, CA 94063-2478, USA
| | - Daniel Rohrer
- Bristol-Myers Squibb, 700 Bay Road, Redwood City, CA 94063-2478, USA
| | - Nils Lonberg
- Bristol-Myers Squibb, 700 Bay Road, Redwood City, CA 94063-2478, USA
| | - Thomas H Oguin
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham NC 27703, USA
| | - Gregory D Sempowski
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham NC 27703, USA
| | - Thomas B Kepler
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Masayuki Kuraoka
- Department of Immunology, Duke University, Durham, NC 27710, USA
| | - Daniel Lingwood
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA.
| | - Aaron G Schmidt
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
33
|
Mahendra A, Haque A, Prabakaran P, Mackness BC, Fuller TP, Liu X, Kathuria SV, Wang YH, Amatya N, Yu X, Hopke J, Hoffmann D, Bric-Furlong E, Zhang N, Cho HS, Zhang R, Sancho J, Saleh J, Rao SP, Wendt M, Chowdhury PS. Honing-in antigen-specific cells during antibody discovery: a user-friendly process to mine a deeper repertoire. Commun Biol 2022; 5:1157. [DOI: 10.1038/s42003-022-04129-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 10/18/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractImmunization based antibody discovery is plagued by the paucity of antigen-specific B cells. Identifying these cells is akin to finding needle in a haystack. Current and emerging technologies while effective, are limited in terms of capturing the antigen-specific repertoire. We report on the bulk purification of antigen-specific B-cells and the benefits it offers to various antibody discovery platforms. Using five different antigens, we show hit rates of 51–88%, compared to about 5% with conventional methods. We also show that this purification is highly efficient with loss of only about 2% antigen specific cells. Furthermore, we compared clones in which cognate chains are preserved with those from display libraries in which chains either from total B cells (TBC) or antigen-specific B cells (AgSC) underwent combinatorial pairing. We found that cognate chain paired clones and combinatorial clones from AgSC library had higher frequency of functional clones and showed greater diversity in sequence and paratope compared to clones from the TBC library. This antigen-specific B-cell selection technique exemplifies a process improvement with reduced cycle time and cost, by removing undesired clones prior to screening and increasing the chance of capturing desirable and rare functional clones in the repertoire.
Collapse
|
34
|
Shen Y, Voigt A, Goranova L, Abed M, Kleiner DE, Maldonado JO, Beach M, Pelayo E, Chiorini JA, Craft WF, Ostrov DA, Ramiya V, Sukumaran S, Tuanyok A, Warner BM, Nguyen CQ. Evidence of a Sjögren's disease-like phenotype following COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.10.20.22281265. [PMID: 36324812 PMCID: PMC9628191 DOI: 10.1101/2022.10.20.22281265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Objectives Sjögren's Disease (SjD) is a chronic and systemic autoimmune disease characterized by lymphocytic infiltration and the development of dry eyes and dry mouth resulting from the secretory dysfunction of the exocrine glands. SARS-CoV-2 may trigger the development or progression of autoimmune diseases, as evidenced by increased autoantibodies in patients and the presentation of cardinal symptoms of SjD. The objective of the study was to determine whether SARS-CoV-2 induces the signature clinical symptoms of SjD. Methods The ACE2-transgenic mice were infected with SARS-CoV-2. SJD profiling was conducted. COVID-19 patients' sera were examined for autoantibodies. Clinical evaluations of convalescent COVID-19 subjects, including minor salivary gland (MSG) biopsies, were collected. Lastly, monoclonal antibodies generated from single B cells of patients were interrogated for ACE2/spike inhibition and nuclear antigens. Results Mice infected with the virus showed a decreased saliva flow rate, elevated antinuclear antibodies (ANAs) with anti-SSB/La, and lymphocyte infiltration in the lacrimal and salivary glands. Sera of COVID-19 patients showed an increase in ANA, anti-SSA/Ro52, and anti-SSB/La. The male patients showed elevated levels of anti-SSA/Ro52 compared to female patients, and female patients had more diverse ANA patterns. Minor salivary gland biopsies of convalescent COVID-19 subjects showed focal lymphocytic infiltrates in four of six subjects, and 2 of 6 subjects had focus scores >2. Lastly, we found monoclonal antibodies produced in recovered patients can both block ACE2/spike interaction and recognize nuclear antigens. Conclusion Overall, our study shows a direct association between SARS-CoV-2 and SjD. Hallmark features of SjD salivary glands were histologically indistinguishable from convalescent COVID-19 subjects. The results potentially implicate that SARS-CoV-2 could be an environmental trigger for SjD. Key Messages What is already known about this subject?SAR-CoV-2 has a tropism for the salivary glands. However, whether the virus can induce clinical phenotypes of Sjögren's disease is unknown.What does this study add?Mice infected with SAR-CoV-2 showed loss of secretory function, elevated autoantibodies, and lymphocyte infiltration in glands.COVID-19 patients showed an increase in autoantibodies. Monoclonal antibodies produced in recovered patients can block ACE2/spike interaction and recognize nuclear antigens.Minor salivary gland biopsies of some convalescent subjects showed focal lymphocytic infiltrates with focus scores.How might this impact on clinical practice or future developments?Our data provide strong evidence for the role of SARS-CoV-2 in inducing Sjögren's disease-like phenotypes.Our work has implications for how patients will be diagnosed and treated effectively.
Collapse
Affiliation(s)
- Yiran Shen
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Alexandria Voigt
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Laura Goranova
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Mehdi Abed
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland
| | - David E Kleiner
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Jose O Maldonado
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland
- AAV Biology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland, USA
| | - Margaret Beach
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland
| | - Eileen Pelayo
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland
| | - John A Chiorini
- AAV Biology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland, USA
| | - William F Craft
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - David A Ostrov
- Department of Pathology, Immunology & Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Vijay Ramiya
- LifeSouth Community Blood Centers, Gainesville Fl
| | | | - Apichai Tuanyok
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Blake M Warner
- Salivary Disorder Unit, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland
| | - Cuong Q Nguyen
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
- Department of Oral Biology, College of Dentistry
- Center of Orphaned Autoimmune Diseases, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
35
|
Tsai CM, Caldera JR, Hajam IA, Chiang AWT, Tsai CH, Li H, Díez ML, Gonzalez C, Trieu D, Martins GA, Underhill DM, Arditi M, Lewis NE, Liu GY. Non-protective immune imprint underlies failure of Staphylococcus aureus IsdB vaccine. Cell Host Microbe 2022; 30:1163-1172.e6. [PMID: 35803276 PMCID: PMC9378590 DOI: 10.1016/j.chom.2022.06.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/10/2022] [Accepted: 06/09/2022] [Indexed: 12/25/2022]
Abstract
Humans frequently encounter Staphylococcus aureus (SA) throughout life. Animal studies have yielded SA candidate vaccines, yet all human SA vaccine trials have failed. One notable vaccine "failure" targeted IsdB, critical for host iron acquisition. We explored a fundamental difference between humans and laboratory animals-natural SA exposure. Recapitulating the failed phase III IsdB vaccine trial, mice previously infected with SA do not mount protective antibody responses to vaccination, unlike naive animals. Non-protective antibodies exhibit increased α2,3 sialylation that blunts opsonophagocytosis and preferentially targets a non-protective IsdB domain. IsdB vaccination of SA-infected mice recalls non-neutralizing humoral responses, further reducing vaccine efficacy through direct antibody competition. IsdB vaccine interference was overcome by immunization against the IsdB heme-binding domain. Purified human IsdB-specific antibodies also blunt IsdB passive immunization, and additional SA vaccines are susceptible to SA pre-exposure. Thus, failed anti-SA immunization trials could be explained by non-protective imprint from prior host-SA interaction.
Collapse
Affiliation(s)
- Chih-Ming Tsai
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
| | - J R Caldera
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Irshad A Hajam
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
| | - Austin W T Chiang
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
| | - Chih-Hsiung Tsai
- Department of Life Sciences, National Cheng Kung University, Tainan 701, Taiwan
| | - Haining Li
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA
| | - María Lázaro Díez
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
| | - Cesia Gonzalez
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
| | - Desmond Trieu
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA
| | - Gislâine A Martins
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - David M Underhill
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Moshe Arditi
- Division of Pediatric Infectious Diseases, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA
| | - George Y Liu
- Department of Pediatrics, University of California, San Diego, San Diego, CA 92093, USA; Division of Infectious Diseases, Rady Children's Hospital, San Diego, CA 92123, USA.
| |
Collapse
|
36
|
T-Cell Receptor Repertoire Sequencing and Its Applications: Focus on Infectious Diseases and Cancer. Int J Mol Sci 2022; 23:ijms23158590. [PMID: 35955721 PMCID: PMC9369427 DOI: 10.3390/ijms23158590] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 11/17/2022] Open
Abstract
The immune system is a dynamic feature of each individual and a footprint of our unique internal and external exposures. Indeed, the type and level of exposure to physical and biological agents shape the development and behavior of this complex and diffuse system. Many pathological conditions depend on how our immune system responds or does not respond to a pathogen or a disease or on how the regulation of immunity is altered by the disease itself. T-cells are important players in adaptive immunity and, together with B-cells, define specificity and monitor the internal and external signals that our organism perceives through its specific receptors, TCRs and BCRs, respectively. Today, high-throughput sequencing (HTS) applied to the TCR repertoire has opened a window of opportunity to disclose T-cell repertoire development and behavior down to the clonal level. Although TCR repertoire sequencing is easily accessible today, it is important to deeply understand the available technologies for choosing the best fit for the specific experimental needs and questions. Here, we provide an updated overview of TCR repertoire sequencing strategies, providers and applications to infectious diseases and cancer to guide researchers’ choice through the multitude of available options. The possibility of extending the TCR repertoire to HLA characterization will be of pivotal importance in the near future to understand how specific HLA genes shape T-cell responses in different pathological contexts and will add a level of comprehension that was unthinkable just a few years ago.
Collapse
|
37
|
Zheng B, Yang Y, Chen L, Wu M, Zhou S. B-Cell Receptor Repertoire Sequencing: Deeper Digging into the Mechanisms and Clinical Aspects of Immune-mediated Diseases. iScience 2022; 25:105002. [PMID: 36157582 PMCID: PMC9494237 DOI: 10.1016/j.isci.2022.105002] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
B cells play an essential role in adaptive immunity and are intimately correlated with pleiotropic immune-mediated diseases. Each B cell occupies a unique B cell receptor (BCR), and all BCRs throughout our body form “BCR repertoire.” With the development of sequencing technology and coupled bioinformatics, accumulating evidence indicates that BCR repertoire largely varies under physiological and pathological conditions. Therefore, comprehensive grasp of BCR repertoire will provide new insights into the pathogenesis of immune-mediated diseases and help exploit efficient diagnostic and treatment strategies. In this review, we start with an overview of BCR repertoire and related sequencing technologies and summarize their current applications in immune-mediated diseases. We also underscore the challenges of this emerging field and propose promising future directions in advancing BCR repertoire exploration.
Collapse
Affiliation(s)
- Bohao Zheng
- Wuxi School of Medicine, Jiangnan University, Wuxi, P. R. China
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China
| | - Yuqing Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China
| | - Lin Chen
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China
| | - Mengrui Wu
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, P. R. China
- Corresponding author
| |
Collapse
|
38
|
de Rutte J, Dimatteo R, Archang MM, van Zee M, Koo D, Lee S, Sharrow AC, Krohl PJ, Mellody M, Zhu S, Eichenbaum JV, Kizerwetter M, Udani S, Ha K, Willson RC, Bertozzi AL, Spangler J, Damoiseaux R, Di Carlo D. Suspendable Hydrogel Nanovials for Massively Parallel Single-Cell Functional Analysis and Sorting. ACS NANO 2022; 16:7242-7257. [PMID: 35324146 PMCID: PMC9869715 DOI: 10.1021/acsnano.1c11420] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Techniques to analyze and sort single cells based on functional outputs, such as secreted products, have the potential to transform our understanding of cellular biology as well as accelerate the development of next-generation cell and antibody therapies. However, secreted molecules rapidly diffuse away from cells, and analysis of these products requires specialized equipment and expertise to compartmentalize individual cells and capture their secretions. Herein, we describe methods to fabricate hydrogel-based chemically functionalized microcontainers, which we call nanovials, and demonstrate their use for sorting single viable cells based on their secreted products at high-throughput using only commonly accessible laboratory infrastructure. These nanovials act as solid supports that facilitate attachment of a variety of adherent and suspension cell types, partition uniform aqueous compartments, and capture secreted proteins. Solutions can be exchanged around nanovials to perform fluorescence immunoassays on secreted proteins. Using this platform and commercial flow sorters, we demonstrate high-throughput screening of stably and transiently transfected producer cells based on relative IgG production. Chinese hamster ovary cells sorted based on IgG production regrew and maintained a high secretion phenotype over at least a week, yielding >40% increase in bulk IgG production rates. We also sorted hybridomas and B lymphocytes based on antigen-specific antibody production. Hybridoma cells secreting an antihen egg lysozyme antibody were recovered from background cells, enriching a population of ∼4% prevalence to >90% following sorting. Leveraging the high-speed sorting capabilities of standard sorters, we sorted >1 million events in <1 h. IgG secreting mouse B cells were also sorted and enriched based on antigen-specific binding. Successful sorting of antibody-secreting B cells combined with the ability to perform single-cell RT-PCR to recover sequence information suggests the potential to perform antibody discovery workflows. The reported nanovials can be easily stored and distributed among researchers, democratizing access to high-throughput functional cell screening.
Collapse
Affiliation(s)
- Joseph de Rutte
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Partillion Bioscience Corporation, Los Angeles, CA 90095, USA
| | - Robert Dimatteo
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA 90095, USA
| | - Maani M. Archang
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Mark van Zee
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Doyeon Koo
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Sohyung Lee
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA 90095, USA
| | - Allison C. Sharrow
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Patrick J. Krohl
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Michael Mellody
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Sheldon Zhu
- Partillion Bioscience Corporation, Los Angeles, CA 90095, USA
| | - James V. Eichenbaum
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Monika Kizerwetter
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Shreya Udani
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Kyung Ha
- Department of Mathematics, University of California, Los Angeles, CA 90095, USA
| | - Richard C. Willson
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, USA
| | - Andrea L. Bertozzi
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA
- Department of Mathematics, University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
| | - Jamie Spangler
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21231, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21231, USA
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Robert Damoiseaux
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| | - Dino Di Carlo
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
39
|
Gong S, Gautam S, Coneglio JD, Scinto HB, Ruprecht RM. Antibody Light Chains: Key to Increased Monoclonal Antibody Yields in Expi293 Cells? Antibodies (Basel) 2022; 11:37. [PMID: 35645210 PMCID: PMC9149950 DOI: 10.3390/antib11020037] [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: 03/19/2022] [Revised: 04/19/2022] [Accepted: 05/10/2022] [Indexed: 02/04/2023] Open
Abstract
When constructing isogenic recombinant IgM-IgG pairs, we discovered that μ heavy chains strongly prefer partnering with λ light chains for optimal IgM expression in transiently cotransfected Expi293 cells. When μ chains were paired with κ light chains, IgM yields were low but increased by logs-up to 20,000 X-by using λ chains instead. Switching light chains did not alter epitope specificity. For dimeric IgA2, optimal expression involved pairing with λ chains, whereas light-chain preference varied for other immunoglobulin classes. In summary, recombinant IgM production can be drastically increased by using λ chains, an important finding in the use of IgM for mucosal immunoprophylaxis.
Collapse
Affiliation(s)
- Siqi Gong
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA 70560, USA or (S.G.); (S.G.); (J.D.C.)
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA;
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Seijal Gautam
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA 70560, USA or (S.G.); (S.G.); (J.D.C.)
- Department of Biology, University of Louisiana at Lafayette, Lafayette, LA 70503, USA
| | - Joshua D. Coneglio
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA 70560, USA or (S.G.); (S.G.); (J.D.C.)
| | - Hanna B. Scinto
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA;
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Ruth M. Ruprecht
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA 70560, USA or (S.G.); (S.G.); (J.D.C.)
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA;
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
- Department of Biology, University of Louisiana at Lafayette, Lafayette, LA 70503, USA
| |
Collapse
|
40
|
Sun H, Hu N, Wang J. Application of Microfluidic Technology in Antibody Screening. Biotechnol J 2022; 17:e2100623. [PMID: 35481726 DOI: 10.1002/biot.202100623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/13/2022] [Accepted: 04/23/2022] [Indexed: 11/07/2022]
Abstract
Specific antibodies are widely used in the biomedical field. Current screening methods for specific antibodies mainly involve hybridoma technology and antibody engineering techniques. However, these technologies suffer from tedious screening processes, long preparation periods, high costs, low efficiency, and a degree of automation, which have become a bottleneck for the screening of specific antibodies. To overcome these difficulties, microfluidics has been developed as a promising technology for high-throughput screening and high purity of antibody. In this review, we provide an overview of the recent advances in microfluidic applications for specific antibody screening. In particular, hybridoma technology and four antibody engineering techniques (including phage display, single B cell antibody screening, antibody expression, and cell-free protein synthesis) based on microfluidics have been introduced, challenges, and the future outlook of these technologies are also discussed. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Heng Sun
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Ning Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Jianhua Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
41
|
Huang HY, Liao HY, Chen X, Wang SW, Cheng CW, Shahed-Al-Mahmud M, Liu YM, Mohapatra A, Chen TH, Lo JM, Wu YM, Ma HH, Chang YH, Tsai HY, Chou YC, Hsueh YP, Tsai CY, Huang PY, Chang SY, Chao TL, Kao HC, Tsai YM, Chen YH, Wu CY, Jan JT, Cheng TJR, Lin KI, Ma C, Wong CH. Vaccination with SARS-CoV-2 spike protein lacking glycan shields elicits enhanced protective responses in animal models. Sci Transl Med 2022; 14:eabm0899. [PMID: 35230146 PMCID: PMC9802656 DOI: 10.1126/scitranslmed.abm0899] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
A major challenge to end the pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is to develop a broadly protective vaccine that elicits long-term immunity. As the key immunogen, the viral surface spike (S) protein is frequently mutated, and conserved epitopes are shielded by glycans. Here, we revealed that S protein glycosylation has site-differential effects on viral infectivity. We found that S protein generated by lung epithelial cells has glycoforms associated with increased infectivity. Compared to the fully glycosylated S protein, immunization of S protein with N-glycans trimmed to the mono-GlcNAc-decorated state (SMG) elicited stronger immune responses and better protection for human angiotensin-converting enzyme 2 (hACE2) transgenic mice against variants of concern (VOCs). In addition, a broadly neutralizing monoclonal antibody was identified from SMG-immunized mice that could neutralize wild-type SARS-CoV-2 and VOCs with subpicomolar potency. Together, these results demonstrate that removal of glycan shields to better expose the conserved sequences has the potential to be an effective and simple approach for developing a broadly protective SARS-CoV-2 vaccine.
Collapse
Affiliation(s)
- Han-Yi Huang
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan.,GIP-TRIAD Master’s Program in Agro-Biomedical Science, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Hsin-Yu Liao
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Xiaorui Chen
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Szu-Wen Wang
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan.,Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Cheng-Wei Cheng
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | | | - Yo-Min Liu
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | | | - Ting-Hua Chen
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Jennifer M. Lo
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Min Wu
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Hsiu-Hua Ma
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Hsuan Chang
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan.,Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Ho-Yang Tsai
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan.,Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Yu-Chi Chou
- Biomedical Translation Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Ching-Yen Tsai
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Pau-Yi Huang
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Sui-Yuan Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei 100233, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Tai-Ling Chao
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Han-Chieh Kao
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Ya-Min Tsai
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Yen-Hui Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Chung-Yi Wu
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Jia-Tsrong Jan
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | | | - Kuo-I Lin
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan.,Corresponding author. (C.M.); (K.-I.L.); . (C.-H.W.)
| | - Che Ma
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan.,Corresponding author. (C.M.); (K.-I.L.); . (C.-H.W.)
| | - Chi-Huey Wong
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan.,Department of Chemistry, Scripps Research, La Jolla, CA 92037, USA.,Corresponding author. (C.M.); (K.-I.L.); . (C.-H.W.)
| |
Collapse
|
42
|
Hauser BM, Sangesland M, St Denis KJ, Lam EC, Case JB, Windsor IW, Feldman J, Caradonna TM, Kannegieter T, Diamond MS, Balazs AB, Lingwood D, Schmidt AG. Rationally designed immunogens enable immune focusing following SARS-CoV-2 spike imprinting. Cell Rep 2022; 38:110561. [PMID: 35303475 PMCID: PMC8898741 DOI: 10.1016/j.celrep.2022.110561] [Citation(s) in RCA: 18] [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: 11/02/2021] [Revised: 01/18/2022] [Accepted: 03/02/2022] [Indexed: 01/11/2023] Open
Abstract
Eliciting antibodies to surface-exposed viral glycoproteins can generate protective responses that control and prevent future infections. Targeting conserved sites may reduce the likelihood of viral escape and limit the spread of related viruses with pandemic potential. Here we leverage rational immunogen design to focus humoral responses on conserved epitopes. Using glycan engineering and epitope scaffolding in boosting immunogens, we focus murine serum antibody responses to conserved receptor binding motif (RBM) and receptor binding domain (RBD) epitopes following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike imprinting. Although all engineered immunogens elicit a robust SARS-CoV-2-neutralizing serum response, RBM-focusing immunogens exhibit increased potency against related sarbecoviruses, SARS-CoV, WIV1-CoV, RaTG13-CoV, and SHC014-CoV; structural characterization of representative antibodies defines a conserved epitope. RBM-focused sera confer protection against SARS-CoV-2 challenge. Thus, RBM focusing is a promising strategy to elicit breadth across emerging sarbecoviruses without compromising SARS-CoV-2 protection. These engineering strategies are adaptable to other viral glycoproteins for targeting conserved epitopes.
Collapse
Affiliation(s)
- Blake M Hauser
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Maya Sangesland
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Kerri J St Denis
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Evan C Lam
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - James Brett Case
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ian W Windsor
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Laboratory of Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jared Feldman
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | | | - Ty Kannegieter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Daniel Lingwood
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Aaron G Schmidt
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
43
|
Cavazzoni CB, Hanson BL, Podestà MA, Bechu ED, Clement RL, Zhang H, Daccache J, Reyes-Robles T, Hett EC, Vora KA, Fadeyi OO, Oslund RC, Hazuda DJ, Sage PT. Follicular T Cells Optimize the Germinal Center Response to SARS-CoV-2 Protein Vaccination in Mice. Cell Rep 2022; 38:110399. [PMID: 35139367 PMCID: PMC8806144 DOI: 10.1016/j.celrep.2022.110399] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/18/2021] [Accepted: 01/26/2022] [Indexed: 12/05/2022] Open
Abstract
Follicular helper T (Tfh) cells promote, whereas follicular regulatory T (Tfr) cells restrain, germinal center (GC) reactions. However, the precise roles of these cells in the complex GC reaction remain poorly understood. Here, we perturb Tfh or Tfr cells after SARS-CoV-2 spike protein vaccination in mice. We find that Tfh cells promote the frequency and somatic hypermutation (SHM) of Spike-specific GC B cells and regulate clonal diversity. Tfr cells similarly control SHM and clonal diversity in the GC but do so by limiting clonal competition. In addition, deletion of Tfh or Tfr cells during primary vaccination results in changes in SHM after vaccine boosting. Aged mice, which have altered Tfh and Tfr cells, have lower GC responses, presenting a bimodal distribution of SHM. Together, these data demonstrate that GC responses to SARS-CoV-2 spike protein vaccines require a fine balance of positive and negative follicular T cell help to optimize humoral immunity.
Collapse
|
44
|
Lim SY, Yamaguchi K, Itakura M, Chikazawa M, Matsuda T, Uchida K. Unique B-1 cells specific for both N-pyrrolated proteins and DNA evolve with apolipoprotein E deficiency. J Biol Chem 2022; 298:101582. [PMID: 35031322 PMCID: PMC8844855 DOI: 10.1016/j.jbc.2022.101582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 11/28/2022] Open
Abstract
Lysine N-pyrrolation, a posttranslational modification, which converts lysine residues to Nε-pyrrole-L-lysine, imparts electronegative properties to proteins, causing them to mimic DNA. Apolipoprotein E (apoE) has been identified as a soluble receptor for pyrrolated proteins (pyrP), and accelerated lysine N-pyrrolation has been observed in apoE-deficient (apoE−/−) hyperlipidemic mice. However, the impact of pyrP accumulation consequent to apoE deficiency on the innate immune response remains unclear. Here, we investigated B-1a cells known to produce germline-encoded immunoglobulin M (IgM) from mice deficient in apoE and identified a particular cell population that specifically produces IgM antibodies against pyrP and DNA. We demonstrated an expansion of B-1a cells involved in IgM production in the peritoneal cavity of apoE−/− mice compared with wild-type mice, consistent with a progressive increase of IgM response in the mouse sera. We found that pyrP exhibited preferential binding to B-1a cells and facilitated the production of IgM. B cell receptor analysis of pyrP-specific B-1a cells showed restricted usage of gene segments selected from the germline gene set; most sequences contained high levels of non-templated-nucleotide additions (N-additions) that could contribute to junctional diversity of B cell receptors. Finally, we report that a subset of monoclonal IgM antibodies against pyrP/DNA established from the apoE−/− mice also contained abundant N-additions. These results suggest that the accumulation of pyrP due to apoE deficiency may influence clonal diversity in the pyrP-specific B cell repertoire. The discovery of these unique B-1a cells for pyrP/DNA provides a key link connecting covalent protein modification, lipoprotein metabolism, and innate immunity.
Collapse
Affiliation(s)
- Sei-Young Lim
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Kosuke Yamaguchi
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Masanori Itakura
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Miho Chikazawa
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Tomonari Matsuda
- Research Center for Environmental Quality Management, Kyoto University, Otsu, Shiga, Japan
| | - Koji Uchida
- Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan; Japan Agency for Medical Research and Development, CREST, Tokyo, Japan.
| |
Collapse
|
45
|
Matochko WL, Nelep C, Chen WC, Grauer S, McFadden K, Wilson V, Oxenoid K. CellCelector™ as a platform in isolating primary B cells for antibody discovery. Antib Ther 2022; 5:11-17. [PMID: 35059561 PMCID: PMC8764991 DOI: 10.1093/abt/tbab030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/10/2021] [Accepted: 12/17/2021] [Indexed: 01/02/2023] Open
Abstract
The most robust strategy in antibody discovery is the use of immunized animals and the ability to isolate and immortalize immune B-cells to hybridoma for further interrogation. However, capturing the full repertoire of an immunized animal is labor intensive, time consuming and limited in throughput. Therefore, techniques to directly mine the antibody repertoire of primary B-cells are of great importance in antibody discovery. In the current study, we present a method to isolate individual antigen-specific primary B-cells using the CellCellector™ single-cell isolation platform from XenoMouse® (XM) immunized with a recombinant therapeutic protein, EGFR. We screened a subset of CD138+ B-cells and identified 238 potential EGFR-specific B-cells from 1189 antibody-secreting cells (ASCs) and isolated 94 by CellCellector. We identified a diverse set of heavy chain complementarity-determining region sequences and cloned and expressed 20 into a standard human immunoglobulin G1 antibody format. We further characterized and identified 13 recombinant antibodies that engage soluble and native forms of EGFR. By extrapolating the method to all 400 000 CD138+ B-cells extracted from one EGFR immunized XM, a potential 1196 unique EGFR-specific antibodies could be discovered. CellCelector allows for interrogating the B-cell pool directly and isolating B-cells specific to the therapeutic target of interest. Furthermore, antibody sequences recovered from isolated B-cells engage the native and recombinant target, demonstrating the CellCellector can serve as a platform in antibody discovery.
Collapse
Affiliation(s)
- Wadim L Matochko
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, Canada
| | - Constantin Nelep
- Marketing and Application Development, ALS Automated Lab Solutions GmbH, Jena, Germany
| | - Weihsu C Chen
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, Canada
| | - Stephanie Grauer
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, Canada
| | - Karyn McFadden
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, Canada
| | - Vicki Wilson
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, Canada
| | - Kirill Oxenoid
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, Canada
| |
Collapse
|
46
|
Zhang YN, Paynter J, Sou C, Fourfouris T, Wang Y, Abraham C, Ngo T, Zhang Y, He L, Zhu J. Mechanism of a COVID-19 nanoparticle vaccine candidate that elicits a broadly neutralizing antibody response to SARS-CoV-2 variants. SCIENCE ADVANCES 2021; 7:eabj3107. [PMID: 34669468 PMCID: PMC8528426 DOI: 10.1126/sciadv.abj3107] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/27/2021] [Indexed: 05/12/2023]
Abstract
Vaccines that induce potent neutralizing antibody (NAb) responses against emerging variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are essential for combating the coronavirus disease 2019 (COVID-19) pandemic. We demonstrated that mouse plasma induced by self-assembling protein nanoparticles (SApNPs) that present 20 rationally designed S2GΔHR2 spikes of the ancestral Wuhan-Hu-1 strain can neutralize the B.1.1.7, B.1.351, P.1, and B.1.617 variants with comparable potency. The adjuvant effect on vaccine-induced immunity was investigated by testing 16 formulations for the multilayered I3-01v9 SApNP. Using single-cell sorting, monoclonal antibodies (mAbs) with diverse neutralization breadth and potency were isolated from mice immunized with the receptor binding domain (RBD), S2GΔHR2 spike, and SApNP vaccines. The mechanism of vaccine-induced immunity was examined in the mouse model. Compared with the soluble spike, the I3-01v9 SApNP showed sixfold longer retention, fourfold greater presentation on follicular dendritic cell dendrites, and fivefold stronger germinal center reactions in lymph node follicles.
Collapse
Affiliation(s)
- Yi-Nan Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jennifer Paynter
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Cindy Sou
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tatiana Fourfouris
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ying Wang
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA 19140, USA
- Department of Microbiology and Immunology, Temple University, Philadelphia, PA 19140, USA
| | - Ciril Abraham
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA 19140, USA
| | - Timothy Ngo
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yi Zhang
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA 19140, USA
- Department of Microbiology and Immunology, Temple University, Philadelphia, PA 19140, USA
| | - Linling He
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
47
|
Schardt JS, Pornnoppadol G, Desai AA, Park KS, Zupancic JM, Makowski EK, Smith MD, Chen H, Garcia de Mattos Barbosa M, Cascalho M, Lanigan TM, Moon JJ, Tessier PM. Discovery and characterization of high-affinity, potent SARS-CoV-2 neutralizing antibodies via single B cell screening. Sci Rep 2021; 11:20738. [PMID: 34671080 PMCID: PMC8528929 DOI: 10.1038/s41598-021-99401-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 09/22/2021] [Indexed: 12/17/2022] Open
Abstract
Monoclonal antibodies that target SARS-CoV-2 with high affinity are valuable for a wide range of biomedical applications involving novel coronavirus disease (COVID-19) diagnosis, treatment, and prophylactic intervention. Strategies for the rapid and reliable isolation of these antibodies, especially potent neutralizing antibodies, are critical toward improved COVID-19 response and informed future response to emergent infectious diseases. In this study, single B cell screening was used to interrogate antibody repertoires of immunized mice and isolate antigen-specific IgG1+ memory B cells. Using these methods, high-affinity, potent neutralizing antibodies were identified that target the receptor-binding domain of SARS-CoV-2. Further engineering of the identified molecules to increase valency resulted in enhanced neutralizing activity. Mechanistic investigation revealed that these antibodies compete with ACE2 for binding to the receptor-binding domain of SARS-CoV-2. These antibodies may warrant further development for urgent COVID-19 applications. Overall, these results highlight the potential of single B cell screening for the rapid and reliable identification of high-affinity, potent neutralizing antibodies for infectious disease applications.
Collapse
Affiliation(s)
- John S. Schardt
- grid.214458.e0000000086837370Departments of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Ghasidit Pornnoppadol
- grid.214458.e0000000086837370Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Alec A. Desai
- grid.214458.e0000000086837370Departments of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Kyung Soo Park
- grid.214458.e0000000086837370Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Jennifer M. Zupancic
- grid.214458.e0000000086837370Departments of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Emily K. Makowski
- grid.214458.e0000000086837370Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Matthew D. Smith
- grid.214458.e0000000086837370Departments of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Hongwei Chen
- grid.214458.e0000000086837370Departments of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | | | - Marilia Cascalho
- grid.214458.e0000000086837370Department of Surgery, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109 USA
| | - Thomas M. Lanigan
- grid.214458.e0000000086837370Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI USA
| | - James J. Moon
- grid.214458.e0000000086837370Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA
| | - Peter M. Tessier
- grid.214458.e0000000086837370Departments of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109 USA ,grid.214458.e0000000086837370University of Michigan, North Campus Research Complex, B10-179, 2800 Plymouth Road, Ann Arbor, MI 48109 USA
| |
Collapse
|
48
|
Parallelism of intestinal secretory IgA shapes functional microbial fitness. Nature 2021; 598:657-661. [PMID: 34646015 DOI: 10.1038/s41586-021-03973-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 08/31/2021] [Indexed: 12/19/2022]
Abstract
Dimeric IgA secreted across mucous membranes in response to nonpathogenic taxa of the microbiota accounts for most antibody production in mammals. Diverse binding specificities can be detected within the polyclonal mucosal IgA antibody response1-10, but limited monoclonal hybridomas have been studied to relate antigen specificity or polyreactive binding to functional effects on microbial physiology in vivo11-17. Here we use recombinant dimeric monoclonal IgAs (mIgAs) to finely map the intestinal plasma cell response to microbial colonization with a single microorganism in mice. We identify a range of antigen-specific mIgA molecules targeting defined surface and nonsurface membrane antigens. Secretion of individual dimeric mIgAs targeting different antigens in vivo showed distinct alterations in the function and metabolism of intestinal bacteria, largely through specific binding. Even in cases in which the same microbial antigen is targeted, microbial metabolic alterations differed depending on IgA epitope specificity. By contrast, bacterial surface coating generally reduced motility and limited bile acid toxicity. The overall intestinal IgA response to a single microbe therefore contains parallel components with distinct effects on microbial carbon-source uptake, bacteriophage susceptibility, motility and membrane integrity.
Collapse
|
49
|
Zhang YN, Paynter J, Sou C, Fourfouris T, Wang Y, Abraham C, Ngo T, Zhang Y, He L, Zhu J. Mechanism of a COVID-19 nanoparticle vaccine candidate that elicits a broadly neutralizing antibody response to SARS-CoV-2 variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.03.26.437274. [PMID: 33791704 PMCID: PMC8010731 DOI: 10.1101/2021.03.26.437274] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Vaccines that induce potent neutralizing antibody (NAb) responses against emerging variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are essential for combating the coronavirus disease 2019 (COVID-19) pandemic. We demonstrated that mouse plasma induced by self-assembling protein nanoparticles (SApNPs) that present 20 rationally designed S2GΔHR2 spikes of the ancestral Wuhan-Hu-1 strain can neutralize the B.1.1.7, B.1.351, P.1, and B.1.617 variants with the same potency. The adjuvant effect on vaccine-induced immunity was investigated by testing 16 formulations for the multilayered I3-01v9 SApNP. Using single-cell sorting, monoclonal antibodies (mAbs) with diverse neutralization breadth and potency were isolated from mice immunized with the receptor binding domain (RBD), S2GΔHR2 spike, and SApNP vaccines. The mechanism of vaccine-induced immunity was examined in mice. Compared with the soluble spike, the I3-01v9 SApNP showed 6-fold longer retention, 4-fold greater presentation on follicular dendritic cell dendrites, and 5-fold stronger germinal center reactions in lymph node follicles.
Collapse
Affiliation(s)
- Yi-Nan Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Jennifer Paynter
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Cindy Sou
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Tatiana Fourfouris
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Ying Wang
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, Pennsylvania 19140, USA
- Department of Microbiology and Immunology, Temple University, Philadelphia, Pennsylvania 19140, USA
| | - Ciril Abraham
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, Pennsylvania 19140, USA
| | - Timothy Ngo
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Yi Zhang
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, Pennsylvania 19140, USA
- Department of Microbiology and Immunology, Temple University, Philadelphia, Pennsylvania 19140, USA
| | - Linling He
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California 92037, USA
| |
Collapse
|
50
|
Cavazzoni CB, Bozza VB, Lucas TC, Conde L, Maia B, Mesin L, Schiepers A, Ersching J, Neris RL, Conde JN, Coelho DR, Lima TM, Alvim RG, Castilho LR, de Paula Neto HA, Mohana-Borges R, Assunção-Miranda I, Nobrega A, Victora GD, Vale AM. The immunodominant antibody response to Zika virus NS1 protein is characterized by cross-reactivity to self. J Exp Med 2021; 218:e20210580. [PMID: 34292314 PMCID: PMC8302445 DOI: 10.1084/jem.20210580] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/24/2021] [Accepted: 06/30/2021] [Indexed: 12/20/2022] Open
Abstract
Besides antigen-specific responses to viral antigens, humoral immune response in virus infection can generate polyreactive and autoreactive antibodies. Dengue and Zika virus infections have been linked to antibody-mediated autoimmune disorders, including Guillain-Barré syndrome. A unique feature of flaviviruses is the secretion of nonstructural protein 1 (NS1) by infected cells. NS1 is highly immunogenic, and antibodies targeting NS1 can have both protective and pathogenic roles. In the present study, we investigated the humoral immune response to Zika virus NS1 and found NS1 to be an immunodominant viral antigen associated with the presence of autoreactive antibodies. Through single B cell cultures, we coupled binding assays and BCR sequencing, confirming the immunodominance of NS1. We demonstrate the presence of self-reactive clones in germinal centers after both infection and immunization, some of which present cross-reactivity with NS1. Sequence analysis of anti-NS1 B cell clones showed sequence features associated with pathogenic autoreactive antibodies. Our findings demonstrate NS1 immunodominance at the cellular level as well as a potential role for NS1 in ZIKV-associated autoimmune manifestations.
Collapse
Affiliation(s)
- Cecilia B. Cavazzoni
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY
| | - Vicente B.T. Bozza
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tostes C.V. Lucas
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Conde
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno Maia
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luka Mesin
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY
| | - Ariën Schiepers
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY
| | - Jonatan Ersching
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY
| | - Romulo L.S. Neris
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jonas N. Conde
- Laboratório de Genômica Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diego R. Coelho
- Laboratório de Genômica Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tulio M. Lima
- Programa de Engenharia Química, Laboratório de Engenharia de Cultivos Celulares, Instituto Alberto Luiz Coimbra de Pós-Graduação e Pesquisa de Engenharia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renata G.F. Alvim
- Programa de Engenharia Química, Laboratório de Engenharia de Cultivos Celulares, Instituto Alberto Luiz Coimbra de Pós-Graduação e Pesquisa de Engenharia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leda R. Castilho
- Programa de Engenharia Química, Laboratório de Engenharia de Cultivos Celulares, Instituto Alberto Luiz Coimbra de Pós-Graduação e Pesquisa de Engenharia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Heitor A. de Paula Neto
- Laboratório de Alvos Moleculares, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ronaldo Mohana-Borges
- Laboratório de Genômica Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Iranaia Assunção-Miranda
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alberto Nobrega
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriel D. Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY
| | - Andre M. Vale
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|