1
|
Hillenbrand M, Esslinger C, Seidenberg J, Weber M, Zingg A, Townsend C, Eicher B, Rutkauskaite J, Riese P, Guzman CA, Fischer K, Schmitt S. Fast-Track Discovery of SARS-CoV-2-Neutralizing Antibodies from Human B Cells by Direct Functional Screening. Viruses 2024; 16:339. [PMID: 38543705 PMCID: PMC10975424 DOI: 10.3390/v16030339] [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: 01/30/2024] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 05/23/2024] Open
Abstract
As the COVID-19 pandemic revealed, rapid development of vaccines and therapeutic antibodies are crucial to guarantee a quick return to the status quo of society. In early 2020, we deployed our droplet microfluidic single-cell-based platform DROPZYLLA® for the generation of cognate antibody repertoires of convalescent COVID-19 donors. Discovery of SARS-CoV-2-specific antibodies was performed upon display of antibodies on the surface of HEK293T cells by antigen-specific sorting using binding to the SARS-CoV-2 spike and absence of binding to huACE2 as the sort criteria. This efficiently yielded antibodies within 3-6 weeks, of which up to 100% were neutralizing. One of these, MTX-COVAB, displaying low picomolar neutralization IC50 of SARS-CoV-2 and with a neutralization potency on par with the Regeneron antibodies, was selected for GMP manufacturing and clinical development in June 2020. MTX-COVAB showed strong efficacy in vivo and neutralized all identified clinically relevant variants of SARS-CoV-2 at the time of its selection. MTX-COVAB completed GMP manufacturing by the end of 2020, but clinical development was stopped when the Omicron variant emerged, a variant that proved to be detrimental to all monoclonal antibodies already approved. The present study describes the capabilities of the DROPZYLLA® platform to identify antibodies of high virus-neutralizing capacity rapidly and directly.
Collapse
Affiliation(s)
- Matthias Hillenbrand
- Memo Therapeutics AG, 8952 Schlieren, Switzerland; (M.H.); (M.W.); (A.Z.); (B.E.); (J.R.); (K.F.); (S.S.)
| | - Christoph Esslinger
- Memo Therapeutics AG, 8952 Schlieren, Switzerland; (M.H.); (M.W.); (A.Z.); (B.E.); (J.R.); (K.F.); (S.S.)
| | - Jemima Seidenberg
- Memo Therapeutics AG, 8952 Schlieren, Switzerland; (M.H.); (M.W.); (A.Z.); (B.E.); (J.R.); (K.F.); (S.S.)
| | - Marcel Weber
- Memo Therapeutics AG, 8952 Schlieren, Switzerland; (M.H.); (M.W.); (A.Z.); (B.E.); (J.R.); (K.F.); (S.S.)
| | - Andreas Zingg
- Memo Therapeutics AG, 8952 Schlieren, Switzerland; (M.H.); (M.W.); (A.Z.); (B.E.); (J.R.); (K.F.); (S.S.)
| | - Catherine Townsend
- Memo Therapeutics AG, 8952 Schlieren, Switzerland; (M.H.); (M.W.); (A.Z.); (B.E.); (J.R.); (K.F.); (S.S.)
| | - Barbara Eicher
- Memo Therapeutics AG, 8952 Schlieren, Switzerland; (M.H.); (M.W.); (A.Z.); (B.E.); (J.R.); (K.F.); (S.S.)
| | - Justina Rutkauskaite
- Memo Therapeutics AG, 8952 Schlieren, Switzerland; (M.H.); (M.W.); (A.Z.); (B.E.); (J.R.); (K.F.); (S.S.)
| | - Peggy Riese
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (P.R.); (C.A.G.)
| | - Carlos A. Guzman
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; (P.R.); (C.A.G.)
| | - Karsten Fischer
- Memo Therapeutics AG, 8952 Schlieren, Switzerland; (M.H.); (M.W.); (A.Z.); (B.E.); (J.R.); (K.F.); (S.S.)
| | - Simone Schmitt
- Memo Therapeutics AG, 8952 Schlieren, Switzerland; (M.H.); (M.W.); (A.Z.); (B.E.); (J.R.); (K.F.); (S.S.)
| |
Collapse
|
2
|
Atanasio A, Franklin MC, Kamat V, Hernandez AR, Badithe A, Ben LH, Jones J, Bautista J, Yancopoulos GD, Olson W, Murphy AJ, Sleeman MA, Orengo JM. Targeting immunodominant Bet v 1 epitopes with monoclonal antibodies prevents the birch allergic response. J Allergy Clin Immunol 2022; 149:200-211. [PMID: 34126155 DOI: 10.1016/j.jaci.2021.05.038] [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: 02/01/2021] [Revised: 04/16/2021] [Accepted: 05/14/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND Blocking the major cat allergen, Fel d 1, with mAbs was effective in preventing an acute cat allergic response. OBJECTIVES This study sought to extend the allergen-specific antibody approach and demonstrate that a combination of mAbs targeting Bet v 1, the immunodominant and most abundant allergenic protein in birch pollen, can prevent the birch allergic response. METHODS Bet v 1-specific mAbs, REGN5713, REGN5714, and REGN5715, were isolated using the VelocImmune platform. Surface plasmon resonance, x-ray crystallography, and cryo-electron microscopy determined binding kinetics and structural data. Inhibition of IgE-binding, basophil activation, and mast cell degranulation were assessed via blocking ELISA, flow cytometry, and the passive cutaneous anaphylaxis mouse model. RESULTS REGN5713, REGN5714, and REGN5715 bind with high affinity and noncompetitively to Bet v 1. A cocktail of all 3 antibodies, REGN5713/14/15, blocks IgE binding to Bet v 1 and inhibits Bet v 1- and birch pollen extract-induced basophil activation ex vivo and mast cell degranulation in vivo. Crystal structures of the complex of Bet v 1 with immunoglobulin antigen-binding fragments of REGN5713 or REGN5715 show distinct interaction sites on Bet v 1. Cryo-electron microscopy reveals a planar and roughly symmetrical complex formed by REGN5713/14/15 bound to Bet v 1. CONCLUSIONS These data confirm the immunodominance of Bet v 1 in birch allergy and demonstrate blockade of the birch allergic response with REGN5713/14/15. Structural analyses show simultaneous binding of REGN5713, REGN5714, and REGN5715 with substantial areas of Bet v 1 exposed, suggesting that targeting specific epitopes is sufficient to block the allergic response.
Collapse
Affiliation(s)
| | | | | | | | | | - Li-Hong Ben
- Regeneron Pharmaceuticals, Inc, Tarrytown, NY
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Hansen J, Baum A, Pascal KE, Russo V, Giordano S, Wloga E, Fulton BO, Yan Y, Koon K, Patel K, Chung KM, Hermann A, Ullman E, Cruz J, Rafique A, Huang T, Fairhurst J, Libertiny C, Malbec M, Lee WY, Welsh R, Farr G, Pennington S, Deshpande D, Cheng J, Watty A, Bouffard P, Babb R, Levenkova N, Chen C, Zhang B, Romero Hernandez A, Saotome K, Zhou Y, Franklin M, Sivapalasingam S, Lye DC, Weston S, Logue J, Haupt R, Frieman M, Chen G, Olson W, Murphy AJ, Stahl N, Yancopoulos GD, Kyratsous CA. Studies in humanized mice and convalescent humans yield a SARS-CoV-2 antibody cocktail. Science 2020; 369:1010-1014. [PMID: 32540901 PMCID: PMC7299284 DOI: 10.1126/science.abd0827] [Citation(s) in RCA: 952] [Impact Index Per Article: 238.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 06/11/2020] [Indexed: 12/18/2022]
Abstract
There is an urgent focus on antibodies that target the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral spike and prevent the virus from entering host cells. Hansen et al. generated a large panel of antibodies against the spike protein from humanized mice and recovered patients. From this panel, they identified several neutralizing antibodies, including pairs that do not compete for binding to the receptor binding domain. Baum et al. focused in on four of these antibodies. All four are effective against known spike variants. However, by growing a pseudovirus that expresses the spike in the presence of individual antibodies, the authors were able to select for spike mutants resistant to that antibody. In contrast, escape mutants are not selected when pseudovirus is grown in the presence of pairs of antibodies that either do not compete or only partially compete for binding to the RBD. Such a pair might be used in a therapeutic antibody cocktail. Science, this issue p. 1010, p. 1014 Neutralizing antibodies have become an important tool in treating infectious diseases. Recently, two separate approaches yielded successful antibody treatments for Ebola—one from genetically humanized mice and the other from a human survivor. Here, we describe parallel efforts using both humanized mice and convalescent patients to generate antibodies against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein, which yielded a large collection of fully human antibodies that were characterized for binding, neutralization, and three-dimensional structure. On the basis of these criteria, we selected pairs of highly potent individual antibodies that simultaneously bind the receptor binding domain of the spike protein, thereby providing ideal partners for a therapeutic antibody cocktail that aims to decrease the potential for virus escape mutants that might arise in response to selective pressure from a single-antibody treatment.
Collapse
Affiliation(s)
- Johanna Hansen
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Alina Baum
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | - Vincenzo Russo
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | - Elzbieta Wloga
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | - Ying Yan
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Katrina Koon
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Krunal Patel
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | - Aynur Hermann
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Erica Ullman
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Jonathan Cruz
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | - Tammy Huang
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | | | - Marine Malbec
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Wen-Yi Lee
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Richard Welsh
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Glen Farr
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | | | - Jemmie Cheng
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Anke Watty
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | - Robert Babb
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | - Calvin Chen
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Bojie Zhang
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | - Kei Saotome
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Yi Zhou
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | | | - David Chien Lye
- National Centre for Infectious Diseases, Tan Tock Seng Hospital, Yong Loo Lin School of Medicine, Lee Kong Chian School of Medicine, 16 Jalan Tan Tock Seng, Singapore 308442, Singapore
| | - Stuart Weston
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - James Logue
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Robert Haupt
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Matthew Frieman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Gang Chen
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - William Olson
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | - Neil Stahl
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | | |
Collapse
|
4
|
Kirpach J, Colone A, Bürckert JP, Faison WJ, Dubois ARSX, Sinner R, Reye AL, Muller CP. Detection of a Low Level and Heterogeneous B Cell Immune Response in Peripheral Blood of Acute Borreliosis Patients With High Throughput Sequencing. Front Immunol 2019; 10:1105. [PMID: 31156648 PMCID: PMC6532064 DOI: 10.3389/fimmu.2019.01105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 04/30/2019] [Indexed: 01/08/2023] Open
Abstract
The molecular diagnosis of acute Borreliosis is complicated and better strategies to improve the diagnostic processes are warranted. High Throughput Sequencing (HTS) of human B cell repertoires after e.g., Dengue virus infection or influenza vaccination revealed antigen-associated “CDR3 signatures” which may have the potential to support diagnosis in infectious diseases. The human B cell immune response to Borrelia burgdorferi sensu lato—the causative agent of Borreliosis—has mainly been studied at the antibody level, while less attention has been given to the cellular part of the humoral immune response. There are indications that Borrelia actively influence the B cell immune response and that it is therefore not directly comparable to responses induced by other infections. The main goal of this study was to identify B cell features that could be used to support diagnosis of Borreliosis. Therefore, we characterized the B cell immune response in these patients by combining multicolor flow cytometry, single Borrelia-reactive B cell receptor (BCR) sequencing, and B cell repertoire deep sequencing. Our phenotyping experiments showed, that there is no significant difference between B cell subpopulations of acute Borreliosis patients and controls. BCR sequences from individual epitope-reactive B cells had little in common between each other. HTS showed, however, a higher complementarity determining region 3 (CDR3) amino acid (aa) sequence overlap between samples from different timepoints in patients as compared to controls. This indicates, that HTS is sensitive enough to detect ongoing B cell immune responses in these patients. Although each individual's repertoire was dominated by rather unique clones, clustering of bulk BCR repertoire sequences revealed a higher overlap of IgG BCR repertoire sequences between acute patients than controls. Even if we have identified a few Borrelia-associated CDR3aa sequences, they seem to be rather unique for each patient and therefore not suitable as biomarkers.
Collapse
Affiliation(s)
- Josiane Kirpach
- Vaccinology and B Cell Immunology, Infectious Diseases Research Unit, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Alessia Colone
- Vaccinology and B Cell Immunology, Infectious Diseases Research Unit, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Jean-Philippe Bürckert
- Vaccinology and B Cell Immunology, Infectious Diseases Research Unit, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - William J Faison
- Vaccinology and B Cell Immunology, Infectious Diseases Research Unit, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Axel R S X Dubois
- Vaccinology and B Cell Immunology, Infectious Diseases Research Unit, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Regina Sinner
- Vaccinology and B Cell Immunology, Infectious Diseases Research Unit, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Anna L Reye
- Vaccinology and B Cell Immunology, Infectious Diseases Research Unit, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Claude P Muller
- Vaccinology and B Cell Immunology, Infectious Diseases Research Unit, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
5
|
Jiménez-Saiz R, Ellenbogen Y, Bruton K, Spill P, Sommer DD, Lima H, Waserman S, Patil SU, Shreffler WG, Jordana M. Human BCR analysis of single-sorted, putative IgE + memory B cells in food allergy. J Allergy Clin Immunol 2019; 144:336-339.e6. [PMID: 30959060 DOI: 10.1016/j.jaci.2019.04.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/13/2019] [Accepted: 04/02/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Rodrigo Jiménez-Saiz
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Mass
| | - Yosef Ellenbogen
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Mass
| | - Kelly Bruton
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Paul Spill
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Doron D Sommer
- Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Hermenio Lima
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Susan Waserman
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Sarita U Patil
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Mass
| | - Wayne G Shreffler
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Mass
| | - Manel Jordana
- McMaster Immunology Research Centre (MIRC), Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
6
|
Zhang Q, Yang J, Bautista J, Badithe A, Olson W, Liu Y. Epitope Mapping by HDX-MS Elucidates the Surface Coverage of Antigens Associated with High Blocking Efficiency of Antibodies to Birch Pollen Allergen. Anal Chem 2018; 90:11315-11323. [DOI: 10.1021/acs.analchem.8b01864] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Qian Zhang
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Rd, Tarrytown, New York 10591, United States
| | - Janice Yang
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Rd, Tarrytown, New York 10591, United States
| | - Joannie Bautista
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Rd, Tarrytown, New York 10591, United States
| | - Ashok Badithe
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Rd, Tarrytown, New York 10591, United States
| | - William Olson
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Rd, Tarrytown, New York 10591, United States
| | - Yashu Liu
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Rd, Tarrytown, New York 10591, United States
| |
Collapse
|
7
|
Jia B, McNeil LK, Dupont CD, Tsioris K, Barry RM, Scully IL, Ogunniyi AO, Gonzalez C, Pride MW, Gierahn TM, Liberator PA, Jansen KU, Love JC. Longitudinal multiparameter single-cell analysis of macaques immunized with pneumococcal protein-conjugated or unconjugated polysaccharide vaccines reveals distinct antigen specific memory B cell repertoires. PLoS One 2017; 12:e0183738. [PMID: 28910279 PMCID: PMC5598952 DOI: 10.1371/journal.pone.0183738] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 08/10/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The efficacy of protein-conjugated pneumococcal polysaccharide vaccines has been well characterized for children. The level of protection conferred by unconjugated polysaccharide vaccines remains less clear, particularly for elderly individuals who have had prior antigenic experience through immunization with unconjugated polysaccharide vaccines or natural exposure to Streptococcus pneumoniae. METHODS We compared the magnitude, diversity and genetic biases of antigen-specific memory B cells in two groups of adult cynomolgus macaques that were immunized with a 7-valent conjugated vaccine and boosted after five years with either a 13-valent pneumococcal polysaccharide conjugate vaccine (13vPnC) or a 23-valent unconjugated pneumococcal polysaccharide vaccine (23vPS) using microengraving (a single-cell analysis method) and single-cell RT-PCR. RESULTS Seven days after boosting, the mean frequency of antigen-specific memory B cells was significantly increased in macaques vaccinated with 13vPnC compared to those receiving 23vPS. The 13vPnC-vaccinated macaques also exhibited a more even distribution of antibody specificities to four polysaccharides in the vaccine (PS4, 6B, 14, 23F) that were examined. However, single-cell analysis of the antibody variable region sequences from antigen-specific B cells elicited by unconjugated and conjugated vaccines indicated that both the germline gene segments forming the heavy chains and the average lengths of the Complementary Determining Region 3 (CDR3) were similar. CONCLUSIONS Our results confirm that distinctive differences can manifest between antigen-specific memory B cell repertoires in nonhuman primates immunized with conjugated and unconjugated pneumococcal polysaccharide vaccines. The study also supports the notion that the conjugated vaccines have a favorable profile in terms of both the frequency and breadth of the anamnestic response among antigen-specific memory B cells.
Collapse
Affiliation(s)
- Bin Jia
- Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Lisa K. McNeil
- Pfizer Vaccine Research and Early Development, Pearl River, New York, United States of America
| | - Christopher D. Dupont
- Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Konstantinos Tsioris
- Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Rachel M. Barry
- Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Ingrid L. Scully
- Pfizer Vaccine Research and Early Development, Pearl River, New York, United States of America
| | - Adebola O. Ogunniyi
- Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Christopher Gonzalez
- Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Michael W. Pride
- Pfizer Vaccine Research and Early Development, Pearl River, New York, United States of America
| | - Todd M. Gierahn
- Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Paul A. Liberator
- Pfizer Vaccine Research and Early Development, Pearl River, New York, United States of America
| | - Kathrin U. Jansen
- Pfizer Vaccine Research and Early Development, Pearl River, New York, United States of America
| | - J. Christopher Love
- Koch Institute for Integrative Cancer Research, Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
| |
Collapse
|
8
|
Hansmann L, Han A, Penter L, Liedtke M, Davis MM. Clonal Expansion and Interrelatedness of Distinct B-Lineage Compartments in Multiple Myeloma Bone Marrow. Cancer Immunol Res 2017; 5:744-754. [PMID: 28768640 PMCID: PMC5590392 DOI: 10.1158/2326-6066.cir-17-0012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 04/05/2017] [Accepted: 07/25/2017] [Indexed: 11/16/2022]
Abstract
Multiple myeloma is characterized by the clonal expansion of malignant plasma cells in the bone marrow. But the phenotypic diversity and the contribution of less predominant B-lineage clones to the biology of this disease have been controversial. Here, we asked whether cells bearing the dominant multiple myeloma immunoglobulin rearrangement occupy phenotypic compartments other than that of plasma cells. To accomplish this, we combined 13-parameter FACS index sorting and t-Stochastic Neighbor Embedding (t-SNE) visualization with high-throughput single-cell immunoglobulin sequencing to track selected B-lineage clones across different stages of human B-cell development. As expected, the predominant clones preferentially mapped to aberrant plasma cell compartments, albeit phenotypically altered from wild type. Interestingly, up to 1.2% of cells of the predominant clones colocalized with B-lineage cells of a normal phenotype. In addition, minor clones with distinct immunoglobulin sequences were detected in up to 9% of sequenced cells, but only 2 out of 12 of these clones showed aberrant immune phenotypes. The majority of these minor clones showed intraclonal silent nucleotide differences within the CDR3s and varying frequencies of somatic mutations in the immunoglobulin genes. Therefore, the phenotypic range of multiple myeloma cells in the bone marrow is not confined to aberrant-phenotype plasma cells but extends to low frequencies of normal-phenotype B cells, in line with the recently reported success of B cell-targeting cellular therapies in some patients. The majority of minor clones result from parallel nonmalignant expansion. Cancer Immunol Res; 5(9); 744-54. ©2017 AACR.
Collapse
Affiliation(s)
- Leo Hansmann
- Department of Microbiology and Immunology, Stanford University, Stanford, California. .,Department of Hematology, Oncology, and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Arnold Han
- Department of Microbiology and Immunology, Stanford University, Stanford, California
| | - Livius Penter
- Department of Hematology, Oncology, and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Michaela Liedtke
- Division of Hematology, Department of Medicine, Stanford University, Stanford, California
| | - Mark M Davis
- Department of Microbiology and Immunology, Stanford University, Stanford, California. .,Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, California.,The Howard Hughes Medical Institute, Stanford University, Stanford, California
| |
Collapse
|
9
|
Quan H, Fang L, Pan H, Deng Z, Gao S, Liu O, Wang Y, Hu Y, Fang X, Yao Z, Guo F, Lu R, Xia K, Tang Z. An adaptive immune response driven by mature, antigen-experienced T and B cells within the microenvironment of oral squamous cell carcinoma. Int J Cancer 2016; 138:2952-62. [PMID: 26815146 DOI: 10.1002/ijc.30019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/16/2015] [Accepted: 01/19/2016] [Indexed: 12/15/2022]
Abstract
Lymphocyte infiltrates have been observed in the microenvironment of oral cancer; however, little is known about whether the immune response of the lymphocyte infiltrate affects tumor biology. For a deeper understanding of the role of the infiltrating-lymphocytes in oral squamous cell carcinoma (OSCC), we characterized the lymphocyte infiltrate repertoires and defined their features. Immunohistochemistry revealed considerable T and B cell infiltrates and lymphoid follicles with germinal center-like structures within the tumor microenvironment. Flow cytometry demonstrated that populations of antigen-experienced CD4+ and CD8+ cells were present, as well as an enrichment of regulatory T cells; and T cells expressing programmed death-1 (PD-1) and T cell Ig and mucin protein-3 (Tim-3), indicative of exhaustion, within the tumor microenvironment. Characterization of tumor-infiltrating B cells revealed clear evidence of antigen exposure, in that the cardinal features of an antigen-driven B cell response were present, including somatic mutation, clonal expansion, intraclonal variation and isotype switching. Collectively, our results point to an adaptive immune response occurring within the OSCC microenvironment, which may be sustained by the expression of specific antigens in the tumor.
Collapse
Affiliation(s)
- Hongzhi Quan
- State Key Laboratory of Medical Genetics, School of Life Science, Central South University, Changsha, Hunan, 410013, People's Republic of China.,Research Institution of Stomatology, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, 410078, People's Republic of China.,Department of Oral Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, 410078, People's Republic of China
| | - Liangjuan Fang
- Department of Immunobiology, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, People's Republic of China
| | - Hao Pan
- Research Institution of Stomatology, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, 410078, People's Republic of China
| | - Zhiyuan Deng
- Research Institution of Stomatology, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, 410078, People's Republic of China
| | - Shan Gao
- Research Institution of Stomatology, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, 410078, People's Republic of China.,Department of Molecular Biology, University of Aarhus, Aarhus C, DK-8000, Denmark
| | - Ousheng Liu
- Research Institution of Stomatology, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, 410078, People's Republic of China
| | - Yuehong Wang
- Research Institution of Stomatology, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, 410078, People's Republic of China
| | - Yanjia Hu
- Department of Oral Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, 410078, People's Republic of China
| | - Xiaodan Fang
- Research Institution of Stomatology, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, 410078, People's Republic of China.,Department of Oral Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, 410078, People's Republic of China
| | - Zhigang Yao
- State Key Laboratory of Medical Genetics, School of Life Science, Central South University, Changsha, Hunan, 410013, People's Republic of China.,Department of Oral Pathology, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, 410078, People's Republic of China
| | - Feng Guo
- Department of Oral Maxillofacial Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, People's Republic of China
| | - Ruohuang Lu
- Department of Oral Maxillofacial Surgery, Xiangya Third Hospital, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Kun Xia
- State Key Laboratory of Medical Genetics, School of Life Science, Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Zhangui Tang
- Research Institution of Stomatology, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, 410078, People's Republic of China.,Department of Oral Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Central South University, Changsha, Hunan, 410078, People's Republic of China
| |
Collapse
|
10
|
Tsioris K, Gupta NT, Ogunniyi AO, Zimnisky RM, Qian F, Yao Y, Wang X, Stern JNH, Chari R, Briggs AW, Clouser CR, Vigneault F, Church GM, Garcia MN, Murray KO, Montgomery RR, Kleinstein SH, Love JC. Neutralizing antibodies against West Nile virus identified directly from human B cells by single-cell analysis and next generation sequencing. Integr Biol (Camb) 2015; 7:1587-97. [PMID: 26481611 PMCID: PMC4754972 DOI: 10.1039/c5ib00169b] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
West Nile virus (WNV) infection is an emerging mosquito-borne disease that can lead to severe neurological illness and currently has no available treatment or vaccine. Using microengraving, an integrated single-cell analysis method, we analyzed a cohort of subjects infected with WNV - recently infected and post-convalescent subjects - and efficiently identified four novel WNV neutralizing antibodies. We also assessed the humoral response to WNV on a single-cell and repertoire level by integrating next generation sequencing (NGS) into our analysis. The results from single-cell analysis indicate persistence of WNV-specific memory B cells and antibody-secreting cells in post-convalescent subjects. These cells exhibited class-switched antibody isotypes. Furthermore, the results suggest that the antibody response itself does not predict the clinical severity of the disease (asymptomatic or symptomatic). Using the nucleotide coding sequences for WNV-specific antibodies derived from single cells, we revealed the ontogeny of expanded WNV-specific clones in the repertoires of recently infected subjects through NGS and bioinformatic analysis. This analysis also indicated that the humoral response to WNV did not depend on an anamnestic response, due to an unlikely previous exposure to the virus. The innovative and integrative approach presented here to analyze the evolution of neutralizing antibodies from natural infection on a single-cell and repertoire level can also be applied to vaccine studies, and could potentially aid the development of therapeutic antibodies and our basic understanding of other infectious diseases.
Collapse
Affiliation(s)
- Konstantinos Tsioris
- Department of Chemical Engineering, Koch Institute of Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Bldg. 76-253, Cambridge, MA 02139, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Shah KA, Clark JJ, Goods BA, Politano TJ, Mozdzierz NJ, Zimnisky RM, Leeson RL, Love JC, Love KR. Automated pipeline for rapid production and screening of HIV-specific monoclonal antibodies using pichia pastoris. Biotechnol Bioeng 2015; 112:2624-9. [PMID: 26032261 DOI: 10.1002/bit.25663] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 05/08/2015] [Accepted: 05/26/2015] [Indexed: 12/30/2022]
Abstract
Monoclonal antibodies (mAbs) that bind and neutralize human pathogens have great therapeutic potential. Advances in automated screening and liquid handling have resulted in the ability to discover antigen-specific antibodies either directly from human blood or from various combinatorial libraries (phage, bacteria, or yeast). There remain, however, bottlenecks in the cloning, expression and evaluation of such lead antibodies identified in primary screens that hinder high-throughput screening. As such, "hit-to-lead identification" remains both expensive and time-consuming. By combining the advantages of overlap extension PCR (OE-PCR) and a genetically stable yet easily manipulatable microbial expression host Pichia pastoris, we have developed an automated pipeline for the rapid production and screening of full-length antigen-specific mAbs. Here, we demonstrate the speed, feasibility and cost-effectiveness of our approach by generating several broadly neutralizing antibodies against human immunodeficiency virus (HIV).
Collapse
Affiliation(s)
- Kartik A Shah
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, 02139, Massachusetts
| | - John J Clark
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, 02139, Massachusetts
| | - Brittany A Goods
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, 02139, Massachusetts
| | - Timothy J Politano
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, 02139, Massachusetts
| | - Nicholas J Mozdzierz
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, 02139, Massachusetts
| | - Ross M Zimnisky
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, 02139, Massachusetts
| | - Rachel L Leeson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, 02139, Massachusetts
| | - J Christopher Love
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, 02139, Massachusetts. .,MIT Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, 02139, Massachusetts.
| | - Kerry R Love
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, 02139, Massachusetts. .,MIT Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, 02139, Massachusetts.
| |
Collapse
|
12
|
Patil SU, Ogunniyi AO, Calatroni A, Tadigotla VR, Ruiter B, Ma A, Moon J, Love J, Shreffler WG. Peanut oral immunotherapy transiently expands circulating Ara h 2-specific B cells with a homologous repertoire in unrelated subjects. J Allergy Clin Immunol 2015; 136:125-134.e12. [PMID: 25985925 PMCID: PMC4494892 DOI: 10.1016/j.jaci.2015.03.026] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 02/16/2015] [Accepted: 03/12/2015] [Indexed: 02/02/2023]
Abstract
BACKGROUND Peanut oral immunotherapy (PNOIT) induces persistent tolerance to peanut in a subset of patients and induces specific antibodies that might play a role in clinical protection. However, the contribution of induced antibody clones to clinical tolerance in PNOIT is unknown. OBJECTIVE We hypothesized that PNOIT induces a clonal, allergen-specific B-cell response that could serve as a surrogate for clinical outcomes. METHODS We used a fluorescent Ara h 2 multimer for affinity selection of Ara h 2-specific B cells and subsequent single-cell immunoglobulin amplification. The diversity of related clones was evaluated by means of next-generation sequencing of immunoglobulin heavy chains from circulating memory B cells with 2x250 paired-end sequencing on the Illumina MiSeq platform. RESULTS Expression of class-switched antibodies from Ara h 2-positive cells confirms enrichment for Ara h 2 specificity. PNOIT induces an early and transient expansion of circulating Ara h 2-specific memory B cells that peaks at week 7. Ara h 2-specific sequences from memory cells have rates of nonsilent mutations consistent with affinity maturation. The repertoire of Ara h 2-specific antibodies is oligoclonal. Next-generation sequencing-based repertoire analysis of circulating memory B cells reveals evidence for convergent selection of related sequences in 3 unrelated subjects, suggesting the presence of similar Ara h 2-specific B-cell clones. CONCLUSIONS Using a novel affinity selection approach to identify antigen-specific B cells, we demonstrate that the early PNOIT-induced Ara h 2-specific B-cell receptor repertoire is oligoclonal and somatically hypermutated and shares similar clonal groups among unrelated subjects consistent with convergent selection.
Collapse
Affiliation(s)
- Sarita U. Patil
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Food Allergy Center, Massachusetts General Hospital and MassGeneral Hospital for Children
- Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Cambridge, MA
| | - Adebola O. Ogunniyi
- Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Cambridge, MA
| | | | | | - Bert Ruiter
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Food Allergy Center, Massachusetts General Hospital and MassGeneral Hospital for Children
| | - Alex Ma
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - James Moon
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - J.Christopher Love
- Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Cambridge, MA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard Medical School
| | - Wayne G. Shreffler
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Food Allergy Center, Massachusetts General Hospital and MassGeneral Hospital for Children
| |
Collapse
|
13
|
Pre- and postexposure efficacy of fully human antibodies against Spike protein in a novel humanized mouse model of MERS-CoV infection. Proc Natl Acad Sci U S A 2015; 112:8738-43. [PMID: 26124093 DOI: 10.1073/pnas.1510830112] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Traditional approaches to antimicrobial drug development are poorly suited to combatting the emergence of novel pathogens. Additionally, the lack of small animal models for these infections hinders the in vivo testing of potential therapeutics. Here we demonstrate the use of the VelocImmune technology (a mouse that expresses human antibody-variable heavy chains and κ light chains) alongside the VelociGene technology (which allows for rapid engineering of the mouse genome) to quickly develop and evaluate antibodies against an emerging viral disease. Specifically, we show the rapid generation of fully human neutralizing antibodies against the recently emerged Middle East Respiratory Syndrome coronavirus (MERS-CoV) and development of a humanized mouse model for MERS-CoV infection, which was used to demonstrate the therapeutic efficacy of the isolated antibodies. The VelocImmune and VelociGene technologies are powerful platforms that can be used to rapidly respond to emerging epidemics.
Collapse
|
14
|
Intrathecal BCR transcriptome in multiple sclerosis versus other neuroinflammation: Equally diverse and compartmentalized, but more mutated, biased and overlapping with the proteome. Clin Immunol 2015; 160:211-25. [PMID: 26055752 DOI: 10.1016/j.clim.2015.06.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 05/28/2015] [Accepted: 06/01/2015] [Indexed: 12/22/2022]
Abstract
The mechanisms driving the intrathecal synthesis of IgG in multiple sclerosis (MS) are unknown. We combined high-throughput sequencing of transcribed immunoglobulin heavy-chain variable (IGHV) genes and mass spectrometry to chart the diversity and compartmentalization of IgG-producing B cells in the cerebrospinal fluid (CSF) of MS patients and controls with other neuroinflammatory diseases. In both groups, a few clones dominated the intrathecal IGHV transcriptome. In most MS patients and some controls, dominant transcripts matched the CSF IgG. The IGHV transcripts in CSF of MS patients frequently carried IGHV4 genes and had more replacement mutations compared to controls. In both groups, dominant IGHV transcripts were identified within clusters of clonally related B cells that had identical or related IGHV transcripts in the blood. These findings suggest more pronounced affinity maturation, but an equal degree of diversity and compartmentalization of the intrathecal B-cell response in MS compared to other neuroinflammatory diseases.
Collapse
|
15
|
Eksteen M, Tiller H, Averina M, Heide G, Kjaer M, Ghevaert C, Michaelsen TE, Ihle Ø, Husebekk A, Skogen B, Stuge TB. Characterization of a human platelet antigen-1a-specific monoclonal antibody derived from a B cell from a woman alloimmunized in pregnancy. THE JOURNAL OF IMMUNOLOGY 2015; 194:5751-60. [PMID: 25972474 DOI: 10.4049/jimmunol.1401599] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 04/09/2015] [Indexed: 11/19/2022]
Abstract
Human platelet Ag (HPA)-1a, located on integrin β3, is the main target for alloantibodies responsible for fetal and neonatal alloimmune thrombocytopenia (FNAIT) in the white population. There are ongoing efforts to develop an Ab prophylaxis and therapy to prevent or treat FNAIT. In this study, an mAb specific for HPA-1a, named 26.4, was derived from an immortalized B cell from an alloimmunized woman who had an infant affected by FNAIT. It is the only HPA-1a-specific human mAb with naturally paired H and L chains. Specific binding of mAb 26.4, both native and recombinant forms, to platelets and to purified integrins αIIbβ3 (from platelets) and αVβ3 (from trophoblasts) from HPA-1a(+) donors was demonstrated by flow cytometry and surface plasmon resonance technology, respectively. No binding to HPA-1a(-) platelets or integrins was detected. Moreover, the Ab binds with higher affinity to integrin αVβ3 compared with a second HPA-1a-specific human mAb, B2G1. Further in vitro experimentation demonstrated that mAb 26.4 can opsonize HPA-1a(+) platelets for enhanced phagocytosis by monocytes, inhibit binding of maternal polyclonal anti-HPA-1a Abs, and weakly inhibit aggregation of HPA-1a-heterozygous platelets, the latter with no predicted clinical relevance. Thus, mAb 26.4 is highly specific for HPA-1a and could potentially be explored for use as a prophylactic or therapeutic reagent for FNAIT intervention and as a phenotyping reagent to identify women at risk for immunization.
Collapse
Affiliation(s)
- Mariana Eksteen
- Immunology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø - The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Heidi Tiller
- Immunology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø - The Arctic University of Norway, N-9037 Tromsø, Norway; Department of Obstetrics and Gynecology, University Hospital of North Norway, N-9038 Tromsø, Norway
| | - Maria Averina
- Department of Laboratory Medicine, Diagnostic Clinic, University Hospital of North Norway, N-9038 Tromsø, Norway
| | - Gøril Heide
- Immunology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø - The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Mette Kjaer
- Immunology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø - The Arctic University of Norway, N-9037 Tromsø, Norway; Department of Laboratory Medicine, Diagnostic Clinic, University Hospital of North Norway, N-9038 Tromsø, Norway; Prophylix Pharma AS, Forskningsparken, N-9294 Tromsø, Norway
| | - Cedric Ghevaert
- Department of Haematology, University of Cambridge, Cambridge CB2 0XY, United Kingdom; National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, United Kingdom
| | - Terje E Michaelsen
- The Norwegian Institute of Public Health, N-0403 Oslo, Norway; and School of Pharmacy, University of Oslo, N-0316 Oslo, Norway
| | - Øistein Ihle
- The Norwegian Institute of Public Health, N-0403 Oslo, Norway; and
| | - Anne Husebekk
- Immunology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø - The Arctic University of Norway, N-9037 Tromsø, Norway; Department of Laboratory Medicine, Diagnostic Clinic, University Hospital of North Norway, N-9038 Tromsø, Norway
| | - Bjørn Skogen
- Immunology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø - The Arctic University of Norway, N-9037 Tromsø, Norway; Department of Laboratory Medicine, Diagnostic Clinic, University Hospital of North Norway, N-9038 Tromsø, Norway; Prophylix Pharma AS, Forskningsparken, N-9294 Tromsø, Norway
| | - Tor B Stuge
- Immunology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø - The Arctic University of Norway, N-9037 Tromsø, Norway;
| |
Collapse
|
16
|
Scherer EM, Smith RA, Simonich CA, Niyonzima N, Carter JJ, Galloway DA. Characteristics of memory B cells elicited by a highly efficacious HPV vaccine in subjects with no pre-existing immunity. PLoS Pathog 2014; 10:e1004461. [PMID: 25330199 PMCID: PMC4199765 DOI: 10.1371/journal.ppat.1004461] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/10/2014] [Indexed: 12/25/2022] Open
Abstract
Licensed human papillomavirus (HPV) vaccines provide near complete protection against the types of HPV that most commonly cause anogenital and oropharyngeal cancers (HPV 16 and 18) when administered to individuals naive to these types. These vaccines, like most other prophylactic vaccines, appear to protect by generating antibodies. However, almost nothing is known about the immunological memory that forms following HPV vaccination, which is required for long-term immunity. Here, we have identified and isolated HPV 16-specific memory B cells from female adolescents and young women who received the quadrivalent HPV vaccine in the absence of pre-existing immunity, using fluorescently conjugated HPV 16 pseudoviruses to label antigen receptors on the surface of memory B cells. Antibodies cloned and expressed from these singly sorted HPV 16-pseudovirus labeled memory B cells were predominantly IgG (>IgA>IgM), utilized diverse variable genes, and potently neutralized HPV 16 pseudoviruses in vitro despite possessing only average levels of somatic mutation. These findings suggest that the quadrivalent HPV vaccine provides an excellent model for studying the development of B cell memory; and, in the context of what is known about memory B cells elicited by influenza vaccination/infection, HIV-1 infection, or tetanus toxoid vaccination, indicates that extensive somatic hypermutation is not required to achieve potent vaccine-specific neutralizing antibody responses. There is an urgent need to better understand how to reliably generate effective vaccines, particularly subunit vaccines, as certain pathogens are considered to pose too great of a safety risk to be developed as live, attenuated or killed vaccines (e.g., HIV-1). The human papillomavirus (HPV) vaccines are two of the most effective subunit vaccines ever developed and have continued to show protection against HPV associated disease up to and beyond five years post-vaccination. Moreover, the target population for these vaccines have essentially no pre-existing immunity to the HPV types covered by the vaccine; therefore, these vaccines provide an excellent model for studying the immunity elicited by a highly effective subunit vaccine. As the HPV vaccines, like most vaccines, protect by generating antibodies, we are interested in characterizing the memory B cells elicited by the HPV vaccine. Memory B cells help to sustain antibody levels over time by rapidly differentiating into antibody secreting cells upon pathogen re-exposure. Although previous studies have provided evidence that the HPV vaccines elicit memory B cells, they did not characterize these cells. Here, we have isolated HPV-specific memory B cells from adolescent females and women who received the quadrivalent HPV vaccine and have cloned antibodies from these cells. Importantly, we find that these antibodies potently inhibit HPV and that the memory B cells from which they derive exhibit hallmarks of long-lived memory B cells.
Collapse
Affiliation(s)
- Erin M. Scherer
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Robin A. Smith
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Cassandra A. Simonich
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington, United States of America
| | - Nixon Niyonzima
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington, United States of America
- Uganda Cancer Institute, Kampala, Uganda
| | - Joseph J. Carter
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Denise A. Galloway
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
17
|
Galson JD, Pollard AJ, Trück J, Kelly DF. Studying the antibody repertoire after vaccination: practical applications. Trends Immunol 2014; 35:319-31. [PMID: 24856924 DOI: 10.1016/j.it.2014.04.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 04/17/2014] [Accepted: 04/28/2014] [Indexed: 12/25/2022]
Abstract
Nearly all licensed vaccines have been developed to confer protection against infectious diseases by stimulating the production of antibodies by B cells, but the nature of a successful antibody response has been difficult to capture. Recent advances in next-generation sequencing (NGS) technology have allowed high-resolution characterization of the antibody repertoire, and of the changes that occur following vaccination. These approaches have yielded important insights into the B cell response, and have raised the possibility of using specific antibody sequences as measures of vaccine immunogenicity. Here, we review recent findings based on antibody repertoire sequencing, and discuss potential applications of these new technologies and of the analyses of the increasing volume of antibody sequence data in the context of vaccine development.
Collapse
Affiliation(s)
- Jacob D Galson
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Johannes Trück
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Dominic F Kelly
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
18
|
Ogunniyi AO, Thomas BA, Politano TJ, Varadarajan N, Landais E, Poignard P, Walker BD, Kwon DS, Love JC. Profiling human antibody responses by integrated single-cell analysis. Vaccine 2014; 32:2866-73. [PMID: 24602776 DOI: 10.1016/j.vaccine.2014.02.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Comprehensive characterization of the antigen-specific B cells induced during infections or following vaccination would facilitate the discovery of novel antibodies and inform how interventions shape protective humoral responses. The analysis of human B cells and their antibodies has been performed using flow cytometry to evaluate memory B cells and expanded plasmablasts, while microtechnologies have also provided a useful tool to examine plasmablasts/plasma cells after vaccination. Here we present an integrated analytical platform, using arrays of subnanoliter wells (nanowells), for constructing detailed profiles for human B cells comprising the immunophenotypes of these cells, the distribution of isotypes of the secreted antibodies, the specificity and relative affinity for defined antigens, and for a subset of cells, the genes encoding the heavy and light chains. The approach combines on-chip image cytometry, microengraving, and single-cell RT-PCR. Using clinical samples from HIV-infected subjects, we demonstrate that the method can identify antigen-specific neutralizing antibodies, is compatible with both plasmablasts/plasma cells and activated memory B cells, and is well-suited for characterizing the limited numbers of B cells isolated from tissue biopsies (e.g., colon biopsies). The technology should facilitate detailed analyses of human humoral responses for evaluating vaccines and their ability to raise protective antibody responses across multiple anatomical compartments.
Collapse
Affiliation(s)
- Adebola O Ogunniyi
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Brittany A Thomas
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Timothy J Politano
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Navin Varadarajan
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77204, United States
| | - Elise Landais
- International AIDS Vaccine Initiative, Scripps Research Institute, La Jolla, CA 92037, United States
| | - Pascal Poignard
- International AIDS Vaccine Initiative, Scripps Research Institute, La Jolla, CA 92037, United States
| | - Bruce D Walker
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, United States
| | - Douglas S Kwon
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, United States
| | - J Christopher Love
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States; The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, United States.
| |
Collapse
|
19
|
Molecular and phenotypic abnormalities of B lymphocytes in patients with Wiskott-Aldrich syndrome. J Allergy Clin Immunol 2013; 133:896-9.e4. [PMID: 24210885 DOI: 10.1016/j.jaci.2013.08.050] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 07/16/2013] [Accepted: 08/28/2013] [Indexed: 01/16/2023]
|
20
|
Fang L, Lowther DE, Meizlish ML, Anderson RCE, Bruce JN, Devine L, Huttner AJ, Kleinstein SH, Lee JY, Stern JNH, Yaari G, Lovato L, Cronk KM, O'Connor KC. The immune cell infiltrate populating meningiomas is composed of mature, antigen-experienced T and B cells. Neuro Oncol 2013; 15:1479-90. [PMID: 23978377 DOI: 10.1093/neuonc/not110] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Meningiomas often harbor an immune cell infiltrate that can include substantial numbers of T and B cells. However, their phenotype and characteristics remain undefined. To gain a deeper understanding of the T and B cell repertoire in this tumor, we characterized the immune infiltrate of 28 resected meningiomas representing all grades. METHODS Immunohistochemistry was used to grossly characterize and enumerate infiltrating lymphocytes. A molecular analysis of the immunoglobulin variable region of tumor-infiltrating B cells was used to characterize their antigen experience. Flow cytometry of fresh tissue homogenate and paired peripheral blood lymphocytes was used to identify T cell phenotypes and characterize the T cell repertoire. RESULTS A conspicuous B and T cell infiltrate, primarily clustered in perivascular spaces, was present in the microenvironment of most tumors examined. Characterization of 294 tumor-infiltrating B cells revealed clear evidence of antigen experience, in that the cardinal features of an antigen-driven B cell response were present. Meningiomas harbored populations of antigen-experienced CD4+ and CD8+ memory/effector T cells, regulatory T cells, and T cells expressing the immune checkpoint molecules PD-1 and Tim-3, indicative of exhaustion. All of these phenotypes were considerably enriched relative to their frequency in the circulation. The T cell repertoire in the tumor microenvironment included populations that were not reflected in paired peripheral blood. CONCLUSION The tumor microenvironment of meningiomas often includes postgerminal center B cell populations. These tumors invariably include a selected, antigen-experienced, effector T cell population enriched by those that express markers of an exhausted phenotype.
Collapse
Affiliation(s)
- Liangjuan Fang
- Corresponding Author: Dr. Kevin C. O'Connor, PhD, Yale School of Medicine, 300 George Street, Room 353J, New Haven, CT, USA 06511..
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Detection and isolation of auto-reactive human antibodies from primary B cells. Methods 2013; 64:153-9. [PMID: 23811296 DOI: 10.1016/j.ymeth.2013.06.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 06/13/2013] [Accepted: 06/18/2013] [Indexed: 12/20/2022] Open
Abstract
The isolation of human monoclonal antibodies (hmAb) has emerged as a versatile platform in a wide variety of contexts ranging from vaccinology to therapeutics. In particular, the presence of high titers of circulating auto-antibodies is implicated in the pathology and outcome of autoimmune diseases. Therefore, the molecular characterization of these hmAb provides an avenue to understanding the pathogenesis of autoimmune diseases. Additionally, the phenotype of the auto-reactive B cells may have direct relevance for therapeutic intervention. In this report, we describe a high-throughput single-cell assay, microengraving, for the screening, characterization and isolation of anti-citrullinated protein antibodies (ACPA) from peripheral blood mononuclear cells (PBMC) of rheumatoid arthritis (RA) patients. Stimulated B cells are profiled at the single-cell level in a large array of sub-nanoliter nanowells (∼10(5)), assessing both the phenotype of the cells and their ability to secrete cyclic-citrullinated peptide (CCP)-specific antibodies. Single B cells secreting ACPA are retrieved by automated micromanipulation, and amplification of the immunoglobulin (Ig) heavy and light chains is performed prior to recombinant expression. The methodology offers a simple, rapid and low-cost platform for isolation of auto-reactive antibodies from low numbers of input cells and can be easily adapted for isolation and characterization of auto-reactive antibodies in other autoimmune diseases.
Collapse
|
22
|
Ray A, Amato AA, Bradshaw EM, Felice KJ, DiCapua DB, Goldstein JM, Lundberg IE, Nowak RJ, Ploegh HL, Spooner E, Wu Q, Willis SN, O’Connor KC. Autoantibodies produced at the site of tissue damage provide evidence of humoral autoimmunity in inclusion body myositis. PLoS One 2012; 7:e46709. [PMID: 23071619 PMCID: PMC3465259 DOI: 10.1371/journal.pone.0046709] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 09/01/2012] [Indexed: 01/07/2023] Open
Abstract
Inclusion body myositis (IBM) belongs to a group of muscle diseases known as the inflammatory myopathies. The presence of antibody-secreting plasma cells in IBM muscle implicates the humoral immune response in this disease. However, whether the humoral immune response actively contributes to IBM pathology has not been established. We sought to investigate whether the humoral immune response in IBM both in the periphery and at the site of tissue damage was directed towards self-antigens. Peripheral autoantibodies present in IBM serum but not control serum recognized self-antigens in both muscle tissue and human-derived cell lines. To study the humoral immune response at the site of tissue damage in IBM patients, we isolated single plasma cells directly from IBM-derived muscle tissue sections and from these cells, reconstructed a series of recombinant immunoglobulins (rIgG). These rIgG, each representing a single muscle-associated plasma cell, were examined for reactivity to self-antigens. Both, flow cytometry and immunoblotting revealed that these rIgG recognized antigens expressed by cell lines and in muscle tissue homogenates. Using a mass spectrometry-based approach, Desmin, a major intermediate filament protein, expressed abundantly in muscle tissue, was identified as the target of one IBM muscle-derived rIgG. Collectively, these data support the view that IBM includes a humoral immune response in both the periphery and at the site of tissue damage that is directed towards self-antigens.
Collapse
Affiliation(s)
- Arundhati Ray
- Department of Neurology, Human and Translational Immunology Program, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Anthony A. Amato
- Department of Neurology, Harvard Medical School and Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Elizabeth M. Bradshaw
- Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kevin J. Felice
- Neuromuscular Program, Hospital for Special Care, New Britain, Connecticut, United States of America
- Department of Neurology, University of Connecticut School of Medicine, Farmington, Connecticut, United States of America
| | - Daniel B. DiCapua
- Department of Neurology, Division of Neuromuscular Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Jonathan M. Goldstein
- Department of Neurology, Division of Neuromuscular Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Ingrid E. Lundberg
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital in Solna, Karolinska Institutet, Stockholm, Sweden
| | - Richard J. Nowak
- Department of Neurology, Division of Neuromuscular Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Hidde L. Ploegh
- Whitehead Institute for Biomedical Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Eric Spooner
- Whitehead Institute for Biomedical Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Qian Wu
- Department of Pathology and Laboratory Medicine, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Simon N. Willis
- Department of Neurology, Human and Translational Immunology Program, Yale School of Medicine, New Haven, Connecticut, United States of America
| | - Kevin C. O’Connor
- Department of Neurology, Human and Translational Immunology Program, Yale School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
23
|
Abstract
Epstein-Barr virus infection has been epidemiologically associated with the development of multiple autoimmune diseases, particularly systemic lupus erythematosus and multiple sclerosis. Currently, there is no known mechanism that can account for these associations. The germinal-center (GC) model of EBV infection and persistence proposes that EBV gains access to the memory B cell compartment via GC reactions by driving infected cells to differentiate using the virus-encoded LMP1 and LMP2a proteins, which act as functional homologues of CD40 and the B cell receptor, respectively. The ability of LMP2a, when expressed in mice, to allow escape of autoreactive B cells suggests that it could perform a similar role in infected GC B cells, permitting the survival of potentially pathogenic autoreactive B cells. To test this hypothesis, we cloned and expressed antibodies from EBV(+) and EBV(-) memory B cells present during acute infection and profiled their self- and polyreactivity. We find that EBV does persist within self- and polyreactive B cells but find no evidence that it favors the survival of pathogenic autoreactive B cells. On the contrary, EBV(+) memory B cells express lower levels of self-reactive and especially polyreactive antibodies than their uninfected counterparts do. Our work suggests that EBV has only a modest effect on the GC process, which allows it to access and persist within a subtly unique niche of the memory compartment characterized by relatively low levels of self- and polyreactivity. We suggest that this might reflect an active process where EBV and its human host have coevolved so as to minimize the virus's potential to contribute to autoimmune disease.
Collapse
|
24
|
Cipponi A, Mercier M, Seremet T, Baurain JF, Théate I, van den Oord J, Stas M, Boon T, Coulie PG, van Baren N. Neogenesis of lymphoid structures and antibody responses occur in human melanoma metastases. Cancer Res 2012; 72:3997-4007. [PMID: 22850419 DOI: 10.1158/0008-5472.can-12-1377] [Citation(s) in RCA: 193] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lymphoid neogenesis, or the development of lymphoid structures in nonlymphoid organs, is frequently observed in chronically inflamed tissues, during the course of autoimmune, infectious, and chronic graft rejection diseases, in which a sustained lymphocyte activation occurs in the presence of persistent antigenic stimuli. The presence of such ectopic lymphoid structures has also been reported in primary lung, breast, and germline cancers, but not yet in melanoma. In this study, we observed ectopic lymphoid structures, defined as lymphoid follicles comprising clusters of B lymphocytes and follicular dendritic cells (DC), associated with high endothelial venules (HEV) and clusters of T cells and mature DCs, in 7 of 29 cutaneous metastases from melanoma patients. Some follicles contained germinal centers. In contrast to metastatic lesions, primary melanomas did not host follicles, but many contained HEVs, suggesting an incomplete lymphoid neogenesis. Analysis of the repertoire of rearranged immunoglobulin genes in the B cells of microdissected follicles revealed clonal amplification, somatic mutation and isotype switching, indicating a local antigen-driven B-cell response. Surprisingly, IgA responses were observed despite the nonmucosal location of the follicles. Taken together, our findings show the existence of lymphoid neogenesis in melanoma and suggest that the presence of functional ectopic lymphoid structures in direct contact with the tumor makes the local development of antimelanoma B- and T-cell responses possible.
Collapse
Affiliation(s)
- Arcadi Cipponi
- de Duve Institute, Université catholique de Louvain, Bruxelles, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Expression of a BCR is critical for B-cell development and survival. We have identified 4 patients with agammaglobulinemia and markedly reduced but detectable B cells in the peripheral circulation. These B cells have an unusual phenotype characterized by increased expression of CD19 but no BCR. The cells are positive for CD20, CD22, and CD38, but not for Annexin 5 or activation markers, including CD69, CD83, or CD86. EBV lines derived from these B cells lack functionally rearranged immunoglobulin heavy-chain transcripts, as shown by PCR-rapid amplification of cDNA ends (PCR-RACE). Analysis of BM from 2 of the patients showed a severe reduction in the number of pro-B cells as well as pre-B cells. Functionally rearranged heavy-chain transcripts were identified, indicating that machinery to rearrange immunoglobulin genes was intact. Flow cytometry of B-lineage cells suggested accelerated acquisition of maturation markers in early B-cell precursors and increased phosphorylation of signal transduction molecules. Further, expression of TdT, a molecule that is normally down-regulated by a functional pre-BCR complex, was decreased. We hypothesize that the accelerated maturation, increased expression of CD19, and lack of a BCR were due to the constitutive activation of the BCR signal transduction pathway in these patients.
Collapse
|
26
|
Ex vivo characterization and isolation of rare memory B cells with antigen tetramers. Blood 2011; 118:348-57. [PMID: 21551230 DOI: 10.1182/blood-2011-03-341917] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Studying human antigen-specific memory B cells has been challenging because of low frequencies in peripheral blood, slow proliferation, and lack of antibody secretion. Therefore, most studies have relied on conversion of memory B cells into antibody-secreting cells by in vitro culture. To facilitate direct ex vivo isolation, we generated fluorescent antigen tetramers for characterization of memory B cells by using tetanus toxoid as a model antigen. Brightly labeled memory B cells were identified even 4 years after last immunization, despite low frequencies ranging from 0.01% to 0.11% of class-switched memory B cells. A direct comparison of monomeric to tetrameric antigen labeling demonstrated that a substantial fraction of the B-cell repertoire can be missed when monomeric antigens are used. The specificity of the method was confirmed by antibody reconstruction from single-cell sorted tetramer(+) B cells with single-cell RT-PCR of the B-cell receptor. All antibodies bound to tetanus antigen with high affinity, ranging from 0.23 to 2.2 nM. Furthermore, sequence analysis identified related memory B cell and plasmablast clones isolated more than a year apart. Therefore, antigen tetramers enable specific and sensitive ex vivo characterization of rare memory B cells as well as the production of fully human antibodies.
Collapse
|
27
|
Abstract
Monoclonal antibodies (mAbs) are arguably the most significant class of biologics for use as pharmaceuticals and diagnostics. Many technological concepts exist for the generation and identification of therapeutically relevant mAbs, including the isolation and cloning of immunoglobulin (Ig) encoding genes from single B-lineage cells. This review summarizes various single B cell approaches and describes their use for the discovery of mAbs with potential therapeutic values or in basic research.
Collapse
Affiliation(s)
- Thomas Tiller
- Max Planck Institute for Infection Biology, D-10117 Berlin, Germany.
| |
Collapse
|
28
|
Ebisawa M, Hase K, Takahashi D, Kitamura H, Knoop KA, Williams IR, Ohno H. CCR6hiCD11c(int) B cells promote M-cell differentiation in Peyer's patch. Int Immunol 2011; 23:261-9. [PMID: 21422150 DOI: 10.1093/intimm/dxq478] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
M cells are responsible for uptake of mucosal antigens in Peyer's patches (PPs). Differentiation of M cells is thought to be induced by interactions between follicle-associated epithelium and PP cells; however, it remains elusive what types of immune cells function as M-cell inducers. Here, we attempted to identify the cells that serve as an M-cell inducer in PP. We found that a unique B-cell subset characterized by CCR6(hi)CD11c(int) resided in the subepithelial dome (SED) in mouse PP. CCR6(hi)CD11c(int) B cells showed chemotactic migration in response to CCL20. Furthermore, this unique B-cell subset substantially decreased in PP of CCR6-deficient mice, indicating that the SED localization of CCR6(hi)CD11c(int) B cells is most likely regulated by the CCL20-CCR6 system. Concomitantly, CCR6 deficiency caused remarkable decrement of M cells. Moreover, adoptive transfer of CCR6(hi)CD11c(int) B cells from wild-type mice restored the M-cell decrement in CCR6-deficient mice. Collectively, the spatial regulation of CCR6(hi)CD11c(int) B cells via the CCL20-CCR6 system may play a vital role in M-cell differentiation in mice.
Collapse
Affiliation(s)
- Masashi Ebisawa
- Laboratory for Epithelial Immunobiology, Research Center for Allergy and Immunology, RIKEN, Yokohama, Kanagawa 230-0045, Japan
| | | | | | | | | | | | | |
Collapse
|
29
|
Griffin DO, Holodick NE, Rothstein TL. Human B1 cells in umbilical cord and adult peripheral blood express the novel phenotype CD20+ CD27+ CD43+ CD70-. ACTA ACUST UNITED AC 2011; 208:67-80. [PMID: 21220451 PMCID: PMC3023138 DOI: 10.1084/jem.20101499] [Citation(s) in RCA: 449] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Human B1 cells consist of CD20+CD27+CD43+CD70− cells bearing a skewed B cell receptor repertoire, and are present in umbilical cord and adult peripheral blood. B1 cells differ in many ways from conventional B cells, most prominently in the production of natural immunoglobulin, which is vitally important for protection against pathogens. B1 cells have also been implicated in the pathogenesis of autoimmune dyscrasias and malignant diseases. It has been impossible to accurately study B1 cells during health and illness because the nature of human B1 cells has not been successfully defined. This has produced controversy regarding the existence of human B1 cells. Here, we determined the phenotype of human B1 cells by testing sort-purified B cell fractions for three fundamental B1 cell functions based on mouse studies: spontaneous IgM secretion, efficient T cell stimulation, and tonic intracellular signaling. We found that a small population of CD20+CD27+CD43+ cells present in both umbilical cord and adult peripheral blood fulfilled these criteria and expressed a skewed B cell receptor repertoire. These B cells express little or no surface CD69 and CD70, both of which are markedly up-regulated after activation of CD20+CD27−CD43− (naive) and CD20+CD27+CD43− (memory) B cells. This work identifies human B1 cells as CD20+CD27+CD43+CD70−. We determined that the proportion of B1 cells declines with age, which may contribute to disease susceptibility. Identification of human B1 cells provides a foundation for future studies on the nature and role of these cells in human disease.
Collapse
Affiliation(s)
- Daniel O Griffin
- Elmezzi Graduate School of Molecular Medicine and Center and for Oncology and Cell Biology, the Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | | | | |
Collapse
|
30
|
Dornmair K, Meinl E, Hohlfeld R. Novel approaches for identifying target antigens of autoreactive human B and T cells. Semin Immunopathol 2009; 31:467-77. [PMID: 19763575 PMCID: PMC2845891 DOI: 10.1007/s00281-009-0179-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Accepted: 08/13/2009] [Indexed: 11/09/2022]
Abstract
Antigen-specific immune responses in multiple sclerosis have been studied for decades, but the target antigens of the putatively autoaggressive B and T cells still remain elusive. Here, we summarize recent strategies which are based on the direct analysis of biopsy or autopsy specimens from patients. Since this material is extremely scarce, the experimental methods need to be exceptionally sensitive. We describe technologies to distinguish (auto) aggressive T cells from irrelevant bystander lymphocytes by analyzing clonal expansions in relation to the morphological location of the cells in the tissue lesions. We then discuss approaches to clone matching α- and β-chains of the antigen-specific T cell receptor (TCR) molecules from single T cells. This is necessary because usually, several clones are expanded and are diluted by many irrelevant cells. The matching TCR chains from individual T cells can be resurrected in hybridoma cells which may then be used for antigen searches. We discuss strategies to identify antigens of γδ- and αβ-TCR molecules, such as biochemical methods, candidate antigens, human leukocyte antigen requirements, synthetic peptide, and cDNA libraries. These strategies are tailored to characterize the antigens of the membrane-anchored, low-affinity TCR molecules. The strategies to identify (auto) reactive B cells or immunoglobulin (Ig) molecules are fundamentally different, because Ig molecules are water-soluble and have high affinities. We further discuss proteome-based approaches, techniques that analyze Ig-chains from single B cells, and a repertoire-based method that compares Ig-proteomes and Ig-transcriptomes. The first method detects Ig antigens directly, whereas the latter two methods allow reconstruction of Ig molecules, which can be used for antigen searches.
Collapse
Affiliation(s)
- Klaus Dornmair
- Department of Neuroimmunology, Max-Planck-Institute of Neurobiology, Am Klopferspitz 18, 82152, Martinsried, Germany.
| | | | | |
Collapse
|
31
|
In vitro generation of anti-hepatitis B monoclonal antibodies from a single plasma cell using single-cell RT-PCR and cell-free protein synthesis. J Biosci Bioeng 2009; 109:75-82. [PMID: 20129087 DOI: 10.1016/j.jbiosc.2009.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Revised: 07/07/2009] [Accepted: 07/07/2009] [Indexed: 11/23/2022]
Abstract
Monoclonal antibodies (mAbs) are an effective tool in therapeutics and diagnostics. A novel approach called the single-cell RT-PCR-linked in vitro expression system (SICREX) enables the high-throughput generation and screening of mAbs from single B cells. In this paper, instead of using B cells, cDNAs were synthesized from single plasma cells of an immunized mouse spleen. The light chain (Lc) and the Fd portion of the heavy chain (Hc) genes of each cell were amplified separately and followed by overlapping PCR to add a T7 promoter, a ribosome-binding site, and a T7 terminator. The paired Lc and Hc genes were simultaneously expressed by an Escherichia coli in vitro transcription and translation system followed by ELISA to measure their affinity for the antigen. A Fab fragment with affinity against the antigen was obtained from plasma cells of an immunized mouse with hepatitis B surface antigen (HBsAg).
Collapse
|
32
|
Production of human monoclonal antibodies against Fc(epsilon)RI(alpha) by a method combining in vitro immunization with phage display. Biosci Biotechnol Biochem 2009; 73:1465-9. [PMID: 19584553 DOI: 10.1271/bbb.80640] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
An in vitro immunization protocol using human peripheral blood mononuclear cells (PBMC) was developed to generate human antigen-specific antibodies. Monoclonal antibodies have great potential, and in particular, efficient acquirement of monoclonal antibodies against membrane proteins provides advantages. In this study, we tried to generate a human monoclonal antibody against the high affinity IgE receptor, Fc(epsilon)RI(alpha), using a method combining in vitro immunization and phage display. Heavy and light chain variable region genes were obtained from PBMC immunized in vitro with Fc(epsilon)RI(alpha)-expressed KU812F cells. Subsequently a combined phage antibody library 6 x 10(3) in the size was generated. Antigen-specific phage antibody clones were selected by panning with recombinant Fc(epsilon)RI(alpha) and recombined to produce human IgG format antibodies using CHO cells. The antibodies exhibited specific binding against Fc(epsilon)RI(alpha). These results suggest that one can obtain membrane protein-specific human monoclonal antibodies from a relatively small phage antibody library using in vitro immunized PBMCs.
Collapse
|
33
|
Willis SN, Mallozzi SS, Rodig SJ, Cronk KM, McArdel SL, Caron T, Pinkus GS, Lovato L, Shampain KL, Anderson DE, Anderson RCE, Bruce JN, O'Connor KC. The microenvironment of germ cell tumors harbors a prominent antigen-driven humoral response. THE JOURNAL OF IMMUNOLOGY 2009; 182:3310-7. [PMID: 19234230 DOI: 10.4049/jimmunol.0803424] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Germ cell tumors are a heterogeneous group of neoplasms derived from residual primordial tissue. These tumors are commonly found in the brain, testes, or ovaries, where they are termed germinomas, seminomas, or dysgerminomas, respectively. Like several other tumor types, germ cell tumors often harbor an immune cell infiltrate that can include substantial numbers of B cells. Yet little is known about whether the humoral immune response affects germ cell tumor biology. To gain a deeper understanding of the role B cells play in this tumor family, we characterized the immune cell infiltrate of all three germ cell tumor subtypes and defined the molecular characteristics of the B cell Ag receptor expressed by tumor-associated B cells. Immunohistochemistry revealed a prominent B cell infiltrate in the microenvironment of all tumors examined and clear evidence of extranodal lymphoid follicles with germinal center-like architecture in a subset of specimens. Molecular characterization of the Ig variable region from 320 sequences expressed by germ cell tumor-infiltrating B cells revealed clear evidence of Ag experience, in that the cardinal features of an Ag-driven B cell response were present: significant somatic mutation, isotype switching, and codon insertion/deletion. This characterization also revealed the presence of both B cell clonal expansion and variation, suggesting that local B cell maturation most likely occurs within the tumor microenvironment. In contrast, sequences from control tissues and peripheral blood displayed none of these characteristics. Collectively, these data strongly suggest that an adaptive and specific humoral immune response is occurring within the tumor microenvironment.
Collapse
Affiliation(s)
- Simon N Willis
- Department of Neurology, Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Liao HX, Levesque MC, Nagel A, Dixon A, Zhang R, Walter E, Parks R, Whitesides J, Marshall DJ, Hwang KK, Yang Y, Chen X, Gao F, Munshaw S, Kepler TB, Denny T, Moody MA, Haynes BF. High-throughput isolation of immunoglobulin genes from single human B cells and expression as monoclonal antibodies. J Virol Methods 2009; 158:171-9. [PMID: 19428587 DOI: 10.1016/j.jviromet.2009.02.014] [Citation(s) in RCA: 188] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2008] [Revised: 02/05/2009] [Accepted: 02/10/2009] [Indexed: 12/28/2022]
Abstract
Defining human B cell repertoires to viral pathogens is critical for design of vaccines that induce broadly protective antibodies to infections such as HIV-1 and influenza. Single B cell sorting and cloning of immunoglobulin (Ig) heavy- and light-chain variable regions (V(H) and V(L)) is a powerful technology for defining anti-viral B cell repertoires. However, the Ig-cloning step is time-consuming and prevents high-throughput analysis of the B cell repertoire. Novel linear Ig heavy- and light-chain gene expression cassettes were designed to express Ig V(H) and V(L) genes isolated from sorted single B cells as IgG1 antibody without a cloning step. The cassettes contain all essential elements for transcriptional and translational regulation, including CMV promoter, Ig leader sequences, constant region of IgG1 heavy- or Ig light-chain, poly(A) tail and substitutable V(H) or V(L) genes. The utility of these Ig gene expression cassettes was established using synthetic V(H) or V(L) genes from an anti-HIV-1 gp41 mAb 2F5 as a model system, and validated further using V(H) and V(L) genes isolated from cloned EBV-transformed antibody-producing cell lines. Finally, this strategy was successfully used for rapid production of recombinant influenza mAbs from sorted single human plasmablasts after influenza vaccination. These Ig gene expression cassettes constitute a highly efficient strategy for rapid expression of Ig genes for high-throughput screening and analysis without cloning.
Collapse
Affiliation(s)
- Hua-Xin Liao
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Rohatgi S, Ganju P, Sehgal D. Systematic design and testing of nested (RT-)PCR primers for specific amplification of mouse rearranged/expressed immunoglobulin variable region genes from small number of B cells. J Immunol Methods 2008; 339:205-19. [PMID: 18926828 DOI: 10.1016/j.jim.2008.09.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Revised: 08/12/2008] [Accepted: 09/18/2008] [Indexed: 11/28/2022]
Abstract
The aim of this study was to develop a highly specific and sensitive (RT-)PCR capable of potentially amplifying the rearranged/expressed VH and VL gene belonging to any mouse immunoglobulin V gene family from a single or a small number of B cells. A database of germline immunoglobulin sequences was used to design 112 primers for a nested (RT-)PCR based strategy to cover all VH, VL, JH, JL, CH and CL gene families/genes from C57BL/6 and BALB/c mice. 93.7% of the primers had 4-fold or less, while 71.4% had no degeneracy. The proportions of germline V genes to which the primers bind with no, up to 1 and up to 2 mismatches are 59.7%, 84.1% and 94.9%, respectively. Most but not all V gene family specific primers designed allow amplification of full-length V genes. The nested primers permit PCR amplification of rearranged V genes belonging to all VH and VL gene families from splenocyte genomic DNA. The V gene family-specific nature of the primers was experimentally confirmed for randomly selected 6 VH and 6 Vkappa families, and all Vlambda genes. The broad V gene family coverage of our primer set was experimentally validated by amplifying the rearranged/expressed VH and VL genes from splenocytes and a panel of 38 hybridomas under conditions where primer mixes and genomic DNA or total RNA was used as starting template. We observed no or low-level cross-family priming. Pooled constant region specific primers allowed efficient RT-PCR amplification of H and L chain isotypes. The expressed VH and VL genes belonging to different V gene families RT-PCR amplified from a mixture of hybridomas in a representative manner. We successfully amplified the expressed VH and Vkappa gene from a single hybridoma cell by RT-PCR and from 10-15 microdissected B cells by genomic PCR. This, first of its kind, comprehensive set of highly sensitive and specific nested primers that provide broad V gene family coverage will open up new avenues and opportunities to study various aspects of mouse B cell biology.
Collapse
Affiliation(s)
- Soma Rohatgi
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | | | | |
Collapse
|
36
|
Matsumoto SE, Yamashita M, Katakura Y, Aiba Y, Tomimatsu K, Kabayama S, Teruya K, Shirahata S. A rapid and efficient strategy to generate antigen-specific human monoclonal antibody by in vitro immunization and the phage display method. J Immunol Methods 2008; 332:2-9. [DOI: 10.1016/j.jim.2007.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2007] [Revised: 11/30/2007] [Accepted: 12/05/2007] [Indexed: 11/30/2022]
|
37
|
Generation of human monoclonal antibodies against Propionibacterium acnes by applying the phage display method to human peripheral blood mononuclear cells immunized in vitro. Cytotechnology 2008; 57:169-75. [PMID: 19003162 DOI: 10.1007/s10616-008-9138-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2007] [Accepted: 02/09/2008] [Indexed: 10/22/2022] Open
Abstract
Propionibacterium acnes is a gram-positive, non-spore-forming, rod-shaped bacterium that is often detected in normal human skin flora. P. acnes has been associated with many diseases. In this study, we attempted to generate anti-P. acnes human monoclonal antibodies. A phage antibody library was first generated from human peripheral blood mononuclear cells immunized in vitro with P. acnes using the phage display method, and P. acnes-specific phage antibodies were obtained using solid phase panning. Antigen-specific variable region genes were then amplified and recombined into vectors expressing human IgG antibodies. The results indicated that the recombinant human IgG antibodies exhibited P. acnes-specific binding. This study demonstrates that the combined use of an in vitro immunization protocol and the phage display method enables the generation of human monoclonal antibodies against pathogenic bacteria and toxic antigens.
Collapse
|
38
|
Wang J, Bu D, Zhu X. Immunoglobulin variable region gene analysis to the autoantibody-secreting B cells from tumors in association with paraneoplastic autoimmune multiorgan syndrome. Int J Dermatol 2008; 46:1146-54. [PMID: 17988333 DOI: 10.1111/j.1365-4632.2007.03267.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVES We have studied the role of lymphoproliferative tumors in the pathogenesis of autoimmune and the origin of the autoantibodies in PNP. Objectives of this study is to understand the characteristics of immunoglobulin genes of the B-cells isolated from the tumor which can produce auto-antibody. METHODS Total RNA of the tumor cells was then isolated and the mRNA was reversely transcribed into cDNA. V(H) and V(L) genes were amplified and cloned and their sequences were analyzed. RESULTS 49 V(H) genes (527 to 577 bp) and 36 V(L) genes (445 to 454 bp) sequences were cloned from five tumors. All of the IgV(L) were mostly homologous to IGKV4-01 germ-line gene. The frequency of IgV(H) germ-line gene usage in a decreasing order was IGHV3-23 > IGHV3-9 > IGHV4-31 > IGHV4-59. There was more nucleotide changes occurred in complementary determining regions (CDR) than those in the framework regions (FR) in the V(H) and V(L) of B cell clones. In both V(H) and V(L), the probability (P) that the number of observed replacement mutations (R) in the CDRs would occur by random chance was low. CONCLUSIONS The clones of B-lymphocytes in the tumors carrying possibly functional rearranged immunoglobulin heavy and light chain genes were isolated and there were high incidences of somatic mutations in CDRs and relatively conserved sequences in framework region.
Collapse
Affiliation(s)
- Jing Wang
- Department of Dermatology, Peking University First Hospital, Beijing, China
| | | | | |
Collapse
|
39
|
Tiller T, Meffre E, Yurasov S, Tsuiji M, Nussenzweig MC, Wardemann H. Efficient generation of monoclonal antibodies from single human B cells by single cell RT-PCR and expression vector cloning. J Immunol Methods 2007; 329:112-24. [PMID: 17996249 DOI: 10.1016/j.jim.2007.09.017] [Citation(s) in RCA: 833] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Revised: 09/25/2007] [Accepted: 09/30/2007] [Indexed: 11/26/2022]
Abstract
We have developed an efficient strategy that combines immunoglobulin (Ig) gene repertoire analysis and Ig reactivity profiling at the single cell level. Based on surface marker expression individual cells at different stages of human B cell development are isolated by fluorescence-activated cell sorting. For each cell Ig heavy and corresponding Ig light chain gene transcripts are amplified by nested RT-PCR and cloned into eukaryotic expression vectors to produce monoclonal human antibodies of the same specificity in vitro. All reactions are performed in 96-well plates and allow cloning of large numbers of Ig genes. The recombinant antibodies are tested for reactivity with diverse self- and non-self antigens and the reactivity profile can be directly linked to the complete Ig heavy and Ig light chain gene sequence information that is obtained as part of the cloning strategy. In summary, our method to clone and express human monoclonal antibodies is unbiased, highly efficient, requires only small cell numbers and the recombinant antibodies allow direct conclusions on the frequency of specific human B cells in a diverse repertoire.
Collapse
Affiliation(s)
- Thomas Tiller
- Max Planck Institute for Infection Biology, D-10117 Berlin, Germany
| | | | | | | | | | | |
Collapse
|
40
|
Souza TA, Stollar BD, Sullivan JL, Luzuriaga K, Thorley-Lawson DA. Influence of EBV on the Peripheral Blood Memory B Cell Compartment. THE JOURNAL OF IMMUNOLOGY 2007; 179:3153-60. [PMID: 17709530 DOI: 10.4049/jimmunol.179.5.3153] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Peripheral blood memory B cells latently infected with EBV bear somatic mutations and are typically isotype switched consistent with being classical Ag-selected memory B cells. In this work, we performed a comparative analysis of the expressed Ig genes between large sets of EBV-infected and uninfected peripheral blood B cells, isolated from the same infectious mononucleosis patients, to determine whether differences exist that could reveal the influence of EBV on the production and maintenance of these cells. We observed that EBV(+) cells on average accumulated more somatic hypermutations than EBV(-) cells. In addition, they had more replacement mutations and a higher replacement-silent ratio of mutations in their CDRs. We also found that EBV occupies a skewed niche within the memory compartment, due to its exclusion from the CD27(+)IgD(+)IgM(+) subset, but this skewing does not affect the overall structure of the compartment. These results indicate that EBV impacts the mutation and selection process of infected cells but that once they enter memory they cannot be distinguished from uninfected cells by host homeostasis mechanisms.
Collapse
Affiliation(s)
- Tatyana A Souza
- Department of Pathology, Jaharis Building, Tufts University School of Medicine, 150 Harrison Avenue, Boston, MA 02111, USA
| | | | | | | | | |
Collapse
|
41
|
Qian Y, Diaz LA, Ye J, Clarke SH. Dissecting the anti-desmoglein autoreactive B cell repertoire in pemphigus vulgaris patients. THE JOURNAL OF IMMUNOLOGY 2007; 178:5982-90. [PMID: 17442983 DOI: 10.4049/jimmunol.178.9.5982] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Pemphigus vulgaris (PV) encompasses two clinical phenotypes, one producing mucosal blisters and the other mucosal and skin lesions (mcPV). The mucosal blister-producing PV variant is characterized by autoantibodies against desmoglein (Dsg)3, whereas mucosal and skin lesion-producing PV is characterized by autoantibodies to Dsg3 and Dsg1. The present study was aimed at disclosing the diversity and clonality of the anti-Dsg3 response, as well as whether anti-Dsg3 B cells are Ag selected. Human-mouse heterohybridomas were generated by fusion of EBV-transformed or freshly isolated PBLs from six PV patients with mouse myeloma cells. A total of 73 anti-Dsg hybridomas (47 IgM and 26 IgG) were isolated. Over 90% are specific for both Dsg1 and Dsg3 indicating extensive cross-reactivity between these responses. V(H) gene segment use by IgM hybridomas is diverse, but is restricted among IgG hybridomas, where the majority uses one of two V(H) genes. V(L) gene segment use was diverse even among IgG hybridomas suggesting that the V(L) is less critical to defining desmoglein specificity. Additionally, the IgG hybridomas were extensively mutated and the distribution and nature of the mutations suggested that they had been Ag selected. We conclude that the potentially pathogenic IgG anti-Dsg response is restricted in V(H) use, is somatically mutated, and is Ag selected.
Collapse
Affiliation(s)
- Ye Qian
- Department of Dermatology, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | |
Collapse
|
42
|
Ghosh S, Huber BT. Clonal diversification in OspA-specific antibodies from peripheral circulation of a chronic Lyme arthritis patient. J Immunol Methods 2007; 321:121-34. [PMID: 17307198 PMCID: PMC1899465 DOI: 10.1016/j.jim.2007.01.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2006] [Accepted: 01/10/2007] [Indexed: 11/18/2022]
Abstract
Chronic, antibiotic treatment-resistant Lyme arthritis develops in a subset of patients following infection with the tick-borne spirochete Borrelia burgdorferi and persists after apparent microbial clearance. IgG responses to Outer Surface Protein (Osp) A, an abundant spirochetal lipoprotein, correlate with both severity and duration of joint inflammation. Characterization of this OspA-directed antibody response is, therefore, important for understanding some of the mechanisms that sustain persistent pathology. Such analyses in Lyme arthritis patients have been previously hampered by relatively small amounts of clinical blood samples, as well as the general intractability and low success rates of B-cell immortalization procedures. Here we describe a robust method for generation of OspA-specific monoclonal antibody fragments from archival cell samples employing a three-step procedure -- isolation of single OspA-specific B-cells, their ex vivo clonal expansion and production of expressed immunoglobulins as single chain variable region fragments (scFvs). Interestingly, two of three scFvs generated from a single patient were of a common clonal origin, additional somatic mutations in the downstream member resulting in a concomitant modulation of antigen binding affinity. Computational docking of OspA into corresponding Fv domains, generated by molecular modeling, reveals subtle binding site differences which could account for the observed alteration in ligand binding. Besides their utility as standards in routine diagnostic assays, being the first described OspA-specific human monoclonal reagents, these scFvs are useful tools for analysis of the anti-OspA repertoire in patients and for identification of putative human mimics of the bacterial protein.
Collapse
MESH Headings
- Amino Acid Sequence
- Antibodies, Bacterial/biosynthesis
- Antibodies, Bacterial/blood
- Antibodies, Bacterial/chemistry
- Antibodies, Bacterial/genetics
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/blood
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/genetics
- Antigens, Surface/chemistry
- Antigens, Surface/genetics
- Antigens, Surface/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Bacterial Outer Membrane Proteins/chemistry
- Bacterial Outer Membrane Proteins/genetics
- Bacterial Outer Membrane Proteins/immunology
- Bacterial Vaccines/chemistry
- Bacterial Vaccines/genetics
- Bacterial Vaccines/immunology
- Borrelia burgdorferi/immunology
- Cell Separation
- Cells, Cultured
- Chronic Disease
- Cloning, Molecular
- Computer Simulation
- DNA Mutational Analysis
- Flow Cytometry
- Humans
- Immunoglobulin Variable Region/biosynthesis
- Immunoglobulin Variable Region/blood
- Immunoglobulin Variable Region/chemistry
- Immunoglobulin Variable Region/genetics
- Lipoproteins/chemistry
- Lipoproteins/genetics
- Lipoproteins/immunology
- Lyme Disease/immunology
- Lyme Disease/metabolism
- Lyme Disease/microbiology
- Lyme Disease/therapy
- Models, Molecular
- Molecular Sequence Data
- Mutation
- Protein Conformation
- Recombinant Proteins/chemistry
- Reverse Transcriptase Polymerase Chain Reaction
- Treatment Failure
Collapse
Affiliation(s)
| | - Brigitte T. Huber
- Corresponding author: Brigitte T. Huber, Department of Pathology, Tufts University School of Medicine, 150 Harrison Avenue, Boston, MA 02111, Phone: 617-636-3989, Fax: 617-636-0449,
| |
Collapse
|
43
|
Culton DA, Nicholas MW, Bunch DO, Zhen QL, Kepler TB, Dooley MA, Mohan C, Nachman PH, Clarke SH. Similar CD19 dysregulation in two autoantibody-associated autoimmune diseases suggests a shared mechanism of B-cell tolerance loss. J Clin Immunol 2006; 27:53-68. [PMID: 17195045 DOI: 10.1007/s10875-006-9051-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2006] [Accepted: 10/06/2006] [Indexed: 10/23/2022]
Abstract
: We report here that dysregulation of CD19, a coreceptor that augments B-cell receptor (BCR) signaling, occurs at two B-cell differentiative stages in patients with systemic lupus erythematosus (SLE) and antineutrophil cytoplasmic autoantibody (ANCA) associated small vessel vasculitis (SVV). The naïve B cells of nearly all SLE and ANCA-SVV patients express approximately 20% less CD19 than healthy control (HC) B cells. In contrast, a subset of memory B cells of some SLE and ANCA-SVV Pts (25-35%) express two to fourfold more CD19 than HC B cells. These CD19(hi) memory B cells are activated and exhibit evidence of antigen selection. Proteome array analysis of 67 autoantigens indicates that CD19(hi) SLE Pts exhibit a distinct autoantibody profile characterized by high levels of antibodies to small nuclear ribonucleoproteins and low levels of antiglomerular autoantibodies. These findings have implications for autoreactive B-cell activation and suggest a shared mechanism of B-cell tolerance loss in these two diseases.
Collapse
Affiliation(s)
- Donna A Culton
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Bradshaw EM, Orihuela A, McArdel SL, Salajegheh M, Amato AA, Hafler DA, Greenberg SA, O'Connor KC. A Local Antigen-Driven Humoral Response Is Present in the Inflammatory Myopathies. THE JOURNAL OF IMMUNOLOGY 2006; 178:547-56. [PMID: 17182595 DOI: 10.4049/jimmunol.178.1.547] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The inflammatory myopathies are putative autoimmune disorders characterized by muscle weakness and the presence of intramuscular inflammatory infiltrates. Although inclusion body myositis and polymyositis have been characterized as cytotoxic CD8(+) T cell-mediated diseases, we recently demonstrated high frequencies of CD138(+) plasma cells in the inflamed muscle tissue of patients with these diseases. To gain a deeper understanding of the role these B cell family members play in the disease pathology, we examined the molecular characteristics of the H chain portion of the Ag receptor. Biopsies of muscle tissue were sectioned and tissue regions and individual cells were isolated through laser capture microdissection. Ig H chain gene transcripts isolated from the sections, regions, and cells were used to determine the variable region gene sequences. Analysis of these sequences revealed clear evidence of affinity maturation in that significant somatic mutation, isotype switching, receptor revision, codon insertion/deletion, and oligoclonal expansion had occurred within the B and plasma cell populations. Moreover, analysis of tissue regions isolated by laser capture microdissection revealed both clonal expansion and variation, suggesting that local B cell maturation occurs within muscle. In contrast, sequences from control muscle tissues and peripheral blood revealed none of these characteristics found in inflammatory myopathy muscle tissue. Collectively, these data demonstrate that Ag drives a B cell Ag-specific response in muscle in patients with dermatomyositis, inclusion body myositis, and polymyositis. These findings highlight the need for a revision of the current paradigm of exclusively T cell-mediated intramuscular Ag-specific autoimmunity in inclusion body myositis and polymyositis.
Collapse
Affiliation(s)
- Elizabeth M Bradshaw
- Department of Neurology, Laboratory of Molecular Immunology, Center for Neurologic Diseases and Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Ghosh S, Seward R, Costello CE, Stollar BD, Huber BT. Autoantibodies from synovial lesions in chronic, antibiotic treatment-resistant Lyme arthritis bind cytokeratin-10. THE JOURNAL OF IMMUNOLOGY 2006; 177:2486-94. [PMID: 16888010 DOI: 10.4049/jimmunol.177.4.2486] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although the causative agent of Lyme disease is definitively known to be the tick-borne spirochete, Borrelia burgdorferi, the etiology of chronic joint inflammation that ensues in a subset of patients remains less well understood. Persistence of arthritis after apparent eradication of the spirochete suggests an autoimmune reaction downstream of the original bacterial infection. We have generated recombinant Ab probes from synovial lesions within affected arthritic joints in an attempt to recapitulate disease-relevant Ag-binding specificities at the site of injury. Using this panel of intra-articular probes, as well as Ab fragments derived from patient peripheral blood, we have identified cytokeratin 10, present in synovial microvascular endothelium, as a target ligand and a putative autoantigen in chronic, antibiotic treatment-resistant Lyme arthritis. Furthermore, there is cross-reactivity between cytokeratin 10 and a prominent B. burgdorferi Ag, outer surface protein A. Release of the self protein in the context of inflammation-induced tissue injury and the resulting in situ response to it could set in motion a feed-forward loop, which amplifies the inflammatory process, thereby rendering it chronic and self-perpetuating, even in the absence of the inciting pathogen.
Collapse
Affiliation(s)
- Srimoyee Ghosh
- Department of Pathology, Tufts University School of Medicine, 150 Harrison Avenue, Boston, MA 02111, USA
| | | | | | | | | |
Collapse
|
46
|
Matsumoto SE, Yamashita M, Katakura Y, Noguchi E, Aiba Y, Ichikawa A, Teruya K, Shirahata S. In vitro immunization can elicit the expansion of diverse repertoire of B cells from peripheral blood mononuclear cells. Cytotechnology 2006; 52:227-33. [PMID: 19002881 DOI: 10.1007/s10616-006-9003-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2006] [Accepted: 03/09/2006] [Indexed: 10/24/2022] Open
Abstract
We previously developed an in vitro immunization (IVI) protocol of human peripheral blood mononuclear cells (PBMC) for generating antigen-specific human antibodies. In order to clarify whether IVI protocolinduces antigen-specific B cell responses in PBMC, we analyzed family gene usage and sequence of the variable region gene of immunoglobulin heavy chain (VH gene) of the antibody produced from the in vitro immunized PBMC. Sequence homology analyses of VH gene demonstrated that a larger repertoire of B cells can be sensitized with mite-extract than with cholera toxin B subunit and rice allergen. Further, antigen-specific B cells were efficiently expanded by using CpG oligodeoxynucleotide as adjuvant. These results suggest that appropriate combination of sensitizing antigen and adjuvant is primarily important for expansion of antigen-specific B cells in IVI protocol.
Collapse
Affiliation(s)
- Shin-Ei Matsumoto
- Graduate School of Systems Life Sciences, Kyushu University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka, 812-8581, Japan
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Kolar GR, Mehta D, Wilson PC, Capra JD. Diversity of the Ig Repertoire is Maintained With Age In Spite of Reduced Germinal Centre Cells in Human Tonsil Lymphoid Tissue. Scand J Immunol 2006; 64:314-24. [PMID: 16918701 DOI: 10.1111/j.1365-3083.2006.01817.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Humans and almost all species studied to date exhibit a decreased responsiveness to immunization and increased autoimmunity with age. While this has been observed clinically for decades, only recently has an understanding of the molecular basis for these changes begun to be appreciated. Studies of the B-cell aspects of these changes in ageing mice and the very few reports in ageing humans have not been conclusive. Here we examine the nucleotide sequence of over 1250 VH transcripts from the tonsils of individuals of various ages for changes to the VH4 immunoglobulin repertoire. An exhaustive examination of VH, DH and JH gene segment utilization revealed a remarkable similarity of the repertoires. The extent of somatic hypermutation was fully maintained or even increased by some measures into the eighth decade of life. However, we found by middle age that the representation of naïve and germinal centre B-cell subpopulations changed relative to total B lymphocytes in the tonsil. While the percentage of naïve and germinal centre B-cell subpopulations changes during the second half of life, these findings suggest that even with advancing age, humans remain capable of generating an extremely diverse Ig repertoire while maintaining a similar spectrum of Ig rearrangements once the germinal centre reaction begins.
Collapse
Affiliation(s)
- G R Kolar
- Program in Molecular Immunogenetics, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104a, USA
| | | | | | | |
Collapse
|
48
|
Ali M, Hitomi K, Nakano H. Generation of monoclonal antibodies using simplified single-cell reverse transcription-polymerase chain reaction and cell-free protein synthesis. J Biosci Bioeng 2006; 101:284-6. [PMID: 16716934 DOI: 10.1263/jbb.101.284] [Citation(s) in RCA: 192] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2005] [Accepted: 12/27/2005] [Indexed: 11/17/2022]
Abstract
The single-step PCR amplification of IgG Light chain (Lc) and Heavy chain (Hc) (Fd portion) from the cDNAs of a single cell was facilitated using a low concentration of cDNA-specific primers with 5' homotags in the presence of a homotag-specific primer. This method was found to be successful in generating a functional antibody with an antigen-binding activity and useful for the high-throughput generation or screening of monoclonal antibodies.
Collapse
Affiliation(s)
- Muhamad Ali
- Laboratory of Molecular Biotechnology, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan
| | | | | |
Collapse
|
49
|
Buschmann C, Fischer C, Ochsenhirt V, Neukirch C, Lackner KJ, von Landenberg P. Generation and characterization of three monoclonal IgM antiphospholipid antibodies recognizing different phospholipid antigens. Ann N Y Acad Sci 2006; 1051:240-54. [PMID: 16126965 DOI: 10.1196/annals.1361.065] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Antiphospholipid antibodies (APLs) might be involved in the pathogenesis of the antiphospholipid syndrome (APS). This study analyzes the structural characteristics of monoclonal APLs derived from patients with this disease. Patient-derived B cells were immortalized using Epstein-Barr virus transformation and subsequent fusion to the myeloma cell line CB-F7. APL-producing hybridomas were cloned to obtain cell lines producing monoclonal APL. DNA encoding the variable region of heavy and light chains of the antibodies was sequenced and analyzed regarding their usage within the V-gene family and the existence of somatic hypermutation. Binding patterns of APL to various phospholipids and beta-2-glycoprotein-I (beta2-GPI) were determined using ELISA, with special regard to beta2-GPI dependency. As a result, three APL-producing hybridoma cell lines from patients with APS were established: JGG9, HVA2, and HLC9. APLs were of the IgM isotype and showed different binding patterns toward phospholipids and beta2-GPI. One of them, JGG9, showed extensive somatic hypermutations in both the CDR3 region and a framework region of the heavy chain. JGG9 bound to cardiolipin in the presence of the protein cofactor beta2-GPI. In contrast, the antibodies HVA2 and HLC9 (which also showed somatic hypermutations in the CDR3 region) presented polyreactivity to several phospholipids-cardiolipin, phosphatidyl-serine, -ethanolamine, -inositol, -choline, and sphingomyelin-but not to beta2-GPI. In conclusion, JGG9 presents a high degree of mutation in the CDR3 and framework region resulting from the deletions of nucleotides, and affects amino acid composition. Polyreactivity and the absence of cofactor dependency present HLC9- and HVA2-like natural antibodies that have no contact with any antigen. Nonetheless, these natural antibodies show somatic hypermutation of the heavy chain, indicating antigen-driven maturation. Regarding the possible role of APL in infection, HVA2 in particular may represent a pathogen-maturated antibody showing cross-reactivity between phospholipids and infectious agents. Further experiments are needed to reveal the functional activity of these antibodies.
Collapse
Affiliation(s)
- Catharina Buschmann
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg University of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | | | | | | | | | | |
Collapse
|
50
|
Ozawa T, Kishi H, Muraguchi A. Amplification and analysis of cDNA generated from a single cell by 5'-RACE: application to isolation of antibody heavy and light chain variable gene sequences from single B cells. Biotechniques 2006; 40:469-70, 472, 474 passim. [PMID: 16629394 DOI: 10.2144/000112123] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The technique of 5'-rapid amplification of cDNA ends (5'-RACE) is widely used to amplify unknown sequences at the 5' end of a messenger RNA (mRNA). However, conventional 5'-RACE is inappropriate for producing cDNAs from a single cell due to the small quantity of mRNA present in one cell. In this study, we report an improved 5'-RACE method that is suitable for generating cDNA from a single cell. In this method, the first-strand cDNA was directly synthesized from a single cell, and both the tailing reaction and second-strand cDNA synthesis were performed in the same tube without purifying the cDNA sample. Using this method, we were able to amplify the cDNA of the immunoglobulin (Ig) variable region gene from more than 50% of single B cells. The amplified cDNA fragment contained a full-length Ig variable region including a 5'-untranslated region, a leader sequence, and an initiation codon. This method may thus be applicable for a comprehensive analysis of the Ig variable genes of the lymphocyte repertoire in humans and animals, thereby contributing to the development of antibody-based therapeutics for infectious diseases.
Collapse
|