1
|
Li X, Liang Y, Zhang Y, Liu Z, Cui L, Xi M, Feng S, Liu X, Zhu Y, Liu S, Li H. Dynamic Immune Response Landscapes of Avian Peripheral Blood Post-Vaccination Against Infectious Bronchitis Virus Infection. Vaccines (Basel) 2025; 13:146. [PMID: 40006693 PMCID: PMC11861738 DOI: 10.3390/vaccines13020146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Despite decades of extensive vaccinations against avian infectious bronchitis virus (IBV) infection, outbreaks caused by constantly emerging variants due to genome recombination between different viral strains, including vaccine strains, occur annually worldwide. The development of novel vaccines with favorable safety and effectiveness is required but is hindered by a limited understanding of vaccination against IBV. Methods: Here, we performed a comprehensive analysis of the in vivo dynamics of peripheral blood mononuclear cells (PBMCs) in specific pathogen-free chickens inoculated with the widely used live attenuated IBV vaccine strain H120 at single-cell level, using high-throughput single-cell transcriptome sequencing (scRNA-seq). Results: High-quality sequencing dataset for four scRNA-seq data containing the transcriptomes of 29,846 individual chicken PBMCs were obtained, defining 22 populations and 7 cell types based on distinct molecular signatures and known markers. Further integrative analysis constructed the time series dynamic cell transition and immune response landscapes within the two weeks post-prime vaccination against IBV. Enhanced crosstalk between antigen-presenting cells and T lymphocytes was revealed as early as four days post-vaccination. The specific immune cell populations and their comprehensive cellular and molecular networks involved in the initiation phase of antiviral adaptive immune responses were elucidated in details. Conclusions: Our study provides a comprehensive view of the dynamic initiation of immune responses in chickens against IBV infection at the cellular and molecular levels, which provides theoretical support and potential solutions for the future rational design of safe and effective vaccines, the augmentation of the efficacy of current vaccines, and the optimization of immune programs.
Collapse
Affiliation(s)
- Xuefeng Li
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, China; (X.L.); (Y.L.); (Y.Z.); (Z.L.); (L.C.)
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (M.X.); (S.F.); (X.L.); (Y.Z.)
| | - Yumeng Liang
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, China; (X.L.); (Y.L.); (Y.Z.); (Z.L.); (L.C.)
| | - Yu Zhang
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, China; (X.L.); (Y.L.); (Y.Z.); (Z.L.); (L.C.)
| | - Zheyi Liu
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, China; (X.L.); (Y.L.); (Y.Z.); (Z.L.); (L.C.)
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (M.X.); (S.F.); (X.L.); (Y.Z.)
| | - Lu Cui
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, China; (X.L.); (Y.L.); (Y.Z.); (Z.L.); (L.C.)
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (M.X.); (S.F.); (X.L.); (Y.Z.)
| | - Miaomiao Xi
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (M.X.); (S.F.); (X.L.); (Y.Z.)
| | - Shufeng Feng
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (M.X.); (S.F.); (X.L.); (Y.Z.)
| | - Xiaoxiao Liu
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (M.X.); (S.F.); (X.L.); (Y.Z.)
| | - Yongxin Zhu
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (M.X.); (S.F.); (X.L.); (Y.Z.)
| | - Shengwang Liu
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, China; (X.L.); (Y.L.); (Y.Z.); (Z.L.); (L.C.)
| | - Hai Li
- Division of Avian Infectious Diseases, State Key Laboratory of Animal Disease Control and Prevention, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, China; (X.L.); (Y.L.); (Y.Z.); (Z.L.); (L.C.)
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (M.X.); (S.F.); (X.L.); (Y.Z.)
| |
Collapse
|
2
|
Heyman B. Antibody feedback regulation. Immunol Rev 2024; 328:126-142. [PMID: 39180190 PMCID: PMC11659925 DOI: 10.1111/imr.13377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2024]
Abstract
Antibodies are able to up- or downregulate antibody responses to the antigen they bind. Two major mechanisms can be distinguished. Suppression is most likely caused by epitope masking and can be induced by all isotypes tested (IgG1, IgG2a, IgG2b, IgG3, IgM, and IgE). Enhancement is often caused by the redistribution of antigen in a favorable way, either for presentation to B cells via follicular dendritic cells (IgM and IgG3) or to CD4+ T cells via dendritic cells (IgE, IgG1, IgG2a, and IgG2b). IgM and IgG3 complexes activate complement and are transported from the marginal zone to follicles by marginal zone B cells expressing complement receptors. IgE-antigen complexes are captured by CD23+ B cells in the blood and transported to follicles, delivered to CD8α+ conventional dendritic cells, and presented to CD4+ T cells. Enhancement of antibody responses by IgG1, IgG2a, and IgG2b in complex with proteins requires activating FcγRs. These immune complexes are captured by dendritic cells and presented to CD4+ T cells, subsequently helping cognate B cells. Endogenous feedback regulation influences the response to booster doses of vaccines and passive administration of anti-RhD antibodies is used to prevent alloimmunization of RhD-negative women carrying RhD-positive fetuses.
Collapse
Affiliation(s)
- Birgitta Heyman
- Department of Medical Biochemistry and MicrobiologyUppsala University, (BMC)UppsalaSweden
| |
Collapse
|
3
|
Daëron M. The function of antibodies. Immunol Rev 2024; 328:113-125. [PMID: 39180466 DOI: 10.1111/imr.13387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2024]
Abstract
Antibodies have multiple biological activities. They can both recognize and act on specific antigens. They can protect against and cause serious diseases, enhance and inhibit antibody responses, enable survival, and threaten life. Which among their many, often antagonistic properties explains that antibodies were selected half a billion years ago and transmitted to mammals across millions of generations? In other words, what is the function of antibodies? Here I examine how their structure endows antibodies with unique cognitive and effector properties that contribute to their multiple biological activities. I show that rather than specific properties, antibodies have large functional repertoires. They have a cognitive repertoire and an effector repertoire that are selected from larger available repertoires, themselves drawn at random from even larger virtual repertoires. These virtual repertoires provide the adaptive immune system with immense, constantly renewed, reservoirs of cognitive and effector functions that can be actualized at any time according to the context. I propose that such a flexibility, which enables living individuals to adapt to a rapidly changing environment, and even deal with an unknown future, may provide a better selective advantage than any particular function.
Collapse
Affiliation(s)
- Marc Daëron
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix Marseille Université-CNRS-Inserm, Marseille, France
- Institut Pasteur-Université Paris Cité, Paris, France
- Institut d'histoire et de philosophie des sciences et des techniques (IHPST), Université Paris 1 Panthéon Sorbonne-CNRS, Paris, France
| |
Collapse
|
4
|
Dvorscek AR, McKenzie CI, Stäheli VC, Ding Z, White J, Fabb SA, Lim L, O'Donnell K, Pitt C, Christ D, Hill DL, Pouton CW, Burnett DL, Brink R, Robinson MJ, Tarlinton DM, Quast I. Conversion of vaccines from low to high immunogenicity by antibodies with epitope complementarity. Immunity 2024; 57:2433-2452.e7. [PMID: 39305904 DOI: 10.1016/j.immuni.2024.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 05/06/2024] [Accepted: 08/27/2024] [Indexed: 10/11/2024]
Abstract
Existing antibodies (Abs) have varied effects on humoral immunity during subsequent infections. Here, we leveraged in vivo systems that allow precise control of antigen-specific Abs and B cells to examine the impact of Ab dose, affinity, and specificity in directing B cell activation and differentiation. Abs competing with the B cell receptor (BCR) epitope showed affinity-dependent suppression. By contrast, Abs targeting a complementary epitope, not overlapping with the BCR, shifted B cell differentiation toward Ab-secreting cells. Such Abs allowed for potent germinal center (GC) responses to otherwise poorly immunogenic sites by promoting antigen capture and presentation by low-affinity B cells. These mechanisms jointly diversified the B cell repertoire by facilitating the recruitment of high- and low-affinity B cells into Ab-secreting cell, GC, and memory B cell fates. Incorporation of small amounts of monoclonal Abs into protein- or mRNA-based vaccines enhanced immunogenicity and facilitated sustained immune responses, with implications for vaccine design and our understanding of protective immunity.
Collapse
Affiliation(s)
- Alexandra R Dvorscek
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Craig I McKenzie
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Vera C Stäheli
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Zhoujie Ding
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Jacqueline White
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Stewart A Fabb
- Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Leonard Lim
- Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Kristy O'Donnell
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Catherine Pitt
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Daniel Christ
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Danika L Hill
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, VIC 3052, Australia
| | - Deborah L Burnett
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia; School of Biomedical Sciences, University of New South Wales, Sydney, NSW 2010, Australia
| | - Robert Brink
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, University of New South Wales, Sydney, NSW 2010, Australia
| | - Marcus J Robinson
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - David M Tarlinton
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia
| | - Isaak Quast
- Department of Immunology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia.
| |
Collapse
|
5
|
Goodwin E, Gibbs JS, Yewdell JW, Eisenlohr LC, Hensley SE. Influenza virus antibodies inhibit antigen-specific de novo B cell responses in mice. J Virol 2024; 98:e0076624. [PMID: 39194245 PMCID: PMC11406888 DOI: 10.1128/jvi.00766-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Antibody responses to influenza vaccines tend to be focused on epitopes encountered during prior influenza exposures, with little production of de novo responses to novel epitopes. To examine the contribution of circulating antibodies to this phenomenon, we passively transferred a hemagglutinin (HA)-specific monoclonal antibody (mAb) into mice before immunizing with whole inactivated virions. The HA mAb inhibited de novo HA-specific antibodies, plasmablasts, germinal center B cells, and memory B cells, while responses to a second antigen in the vaccine, neuraminidase (NA), were uninhibited. The HA mAb potently inhibited de novo antibody responses against epitopes near the HA mAb binding site. The HA mAb also promoted IgG1 class switching, an effect that, unlike the inhibition of HA responses, relied on signaling through Fc-gamma receptors. These studies suggest that circulating antibodies inhibit de novo B cell responses in an antigen-specific manner, which likely contributes to differences in antibody specificities elicited during primary and secondary influenza virus exposures.IMPORTANCEMost humans are exposed to influenza viruses in childhood and then subsequently exposed to antigenically drifted influenza variants later in life. It is unclear if antibodies elicited by earlier influenza virus exposures impact immunity against new influenza virus strains. Here, we used a mouse model to investigate how an anti-hemagglutinin (HA) monoclonal antibody (mAb) affects de novo B cell and antibody responses to the protein targeted by the monoclonal antibody (HA) and a second protein not targeted by the monoclonal antibody [neuraminidase (NA)]. Collectively, our studies suggest that circulating anti-influenza virus antibodies can potently modulate the magnitude and specificity of antibody responses elicited by secondary influenza virus exposures.
Collapse
Affiliation(s)
- Eileen Goodwin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James S. Gibbs
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jonathan W. Yewdell
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Laurence C. Eisenlohr
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Scott E. Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
6
|
Cyster JG, Wilson PC. Antibody modulation of B cell responses-Incorporating positive and negative feedback. Immunity 2024; 57:1466-1481. [PMID: 38986442 PMCID: PMC11257158 DOI: 10.1016/j.immuni.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024]
Abstract
Antibodies are powerful modulators of ongoing and future B cell responses. While the concept of antibody feedback has been appreciated for over a century, the topic has seen a surge in interest due to the evidence that the broadening of antibody responses to SARS-CoV-2 after a third mRNA vaccination is a consequence of antibody feedback. Moreover, the discovery that slow antigen delivery can lead to more robust humoral immunity has put a spotlight on the capacity for early antibodies to augment B cell responses. Here, we review the mechanisms whereby antibody feedback shapes B cell responses, integrating findings in humans and in mouse models. We consider the major influence of epitope masking and the diverse actions of complement and Fc receptors and provide a framework for conceptualizing the ways antigen-specific antibodies may influence B cell responses to any form of antigen, in conditions as diverse as infectious disease, autoimmunity, and cancer.
Collapse
Affiliation(s)
- Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA.
| | - Patrick C Wilson
- Drukier Institute for Children's Health, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Goodwin E, Gibbs JS, Yewdell JW, Eisenlohr LC, Hensley SE. Influenza virus antibodies inhibit antigen-specific de novo B cell responses in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.589218. [PMID: 38659819 PMCID: PMC11042189 DOI: 10.1101/2024.04.12.589218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Antibody responses to influenza vaccines tend to be focused on epitopes encountered during prior influenza exposures, with little production of de novo responses to novel epitopes. To examine the contribution of circulating antibody to this phenomenon, we passively transferred a hemagglutinin (HA)-specific monoclonal antibody (mAb) into mice before immunizing with whole inactivated virions. The HA mAb inhibited de novo HA-specific antibodies, plasmablasts, germinal center B cells, and memory B cells, while responses to a second antigen in the vaccine, neuraminidase (NA), were uninhibited. The HA mAb potently inhibited de novo antibody responses against epitopes near the HA mAb binding site. The HA mAb also promoted IgG1 class switching, an effect that, unlike the inhibition of HA responses, relied on signaling through Fc-gamma receptors. These studies suggest that circulating antibodies inhibit de novo B cell responses in an antigen-specific manner, which likely contributes to differences in antibody specificities elicited during primary and secondary influenza virus exposures.
Collapse
Affiliation(s)
- Eileen Goodwin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - James S. Gibbs
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Jonathan W. Yewdell
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Laurence C. Eisenlohr
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Scott E. Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
8
|
Carpenter MC, Souter SC, Zipkin RJ, Ackerman ME. Current Insights Into K-associated Fetal Anemia and Potential Treatment Strategies for Sensitized Pregnancies. Transfus Med Rev 2024; 38:150779. [PMID: 37926651 PMCID: PMC10856777 DOI: 10.1016/j.tmrv.2023.150779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/21/2023] [Accepted: 09/27/2023] [Indexed: 11/07/2023]
Abstract
K-associated anemic disease of the fetus and newborn (K-ADFN) is a rare but life-threatening disease in which maternal alloantibodies cross the placenta and can mediate an immune attack on fetal red blood cells expressing the K antigen. A considerably more common disease, D-associated hemolytic disease of the fetus and newborn (D-HDFN), can be prophylactically treated using polyclonal α-D antibody preparations. Currently, no such prophylactic treatment exists for K-associated fetal anemia, and disease is usually treated with intrauterine blood transfusions. Here we review current understanding of the biology of K-associated fetal anemia, how the maternal immune system is sensitized to fetal red blood cells, and what is understood about potential mechanisms of prophylactic HDFN interventions. Given the apparent challenges associated with preventing alloimmunization, we highlight novel strategies for treating sensitized mothers to prevent fetal anemia that may hold promise not only for K-mediated disease, but also for other pathogenic alloantibody responses.
Collapse
Affiliation(s)
| | | | | | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA; Geisel School of Medicine at Dartmouth, Lebanon, NH, USA.
| |
Collapse
|
9
|
Inoue T, Shinnakasu R, Kawai C, Yamamoto H, Sakakibara S, Ono C, Itoh Y, Terooatea T, Yamashita K, Okamoto T, Hashii N, Ishii-Watabe A, Butler NS, Matsuura Y, Matsumoto H, Otsuka S, Hiraoka K, Teshima T, Murakami M, Kurosaki T. Antibody feedback contributes to facilitating the development of Omicron-reactive memory B cells in SARS-CoV-2 mRNA vaccinees. J Exp Med 2023; 220:213745. [PMID: 36512034 PMCID: PMC9750191 DOI: 10.1084/jem.20221786] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/16/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022] Open
Abstract
In contrast to a second dose of the SARS-CoV-2 mRNA vaccine, a third dose elicits potent neutralizing activity against the Omicron variant. To address the underlying mechanism for this differential antibody response, we examined spike receptor-binding domain (RBD)-specific memory B cells in vaccinated individuals. Frequency of Omicron-reactive memory B cells increased ∼9 mo after the second vaccine dose. These memory B cells show an altered distribution of epitopes from pre-second memory B cells, presumably due to an antibody feedback mechanism. This hypothesis was tested using mouse models, showing that an addition or a depletion of RBD-induced serum antibodies results in a concomitant increase or decrease, respectively, of Omicron-reactive germinal center (GC) and memory B cells. Our data suggest that pre-generated antibodies modulate the selection of GC and subsequent memory B cells after the second vaccine dose, accumulating more Omicron-reactive memory B cells over time, which contributes to the generation of Omicron-neutralizing antibodies elicited by the third vaccine dose.
Collapse
Affiliation(s)
- Takeshi Inoue
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Ryo Shinnakasu
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of Medical Research Support, Advanced Research Support Center, Ehime University, Ehime, Japan.,Translational Research Center, Ehime University Hospital, Ehime, Japan
| | - Chie Kawai
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Hiromi Yamamoto
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shuhei Sakakibara
- Laboratory of Immune Regulation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Chikako Ono
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Yumi Itoh
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | | | | | - Toru Okamoto
- Institute for Advanced Co-Creation Studies, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Noritaka Hashii
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kanagawa, Japan
| | - Akiko Ishii-Watabe
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, Kanagawa, Japan
| | - Noah S Butler
- Department of Microbiology and Immunology, The University of Iowa, Iowa City, IA, USA
| | - Yoshiharu Matsuura
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Hisatake Matsumoto
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinya Otsuka
- Department of Surgery, National Hospital Organization Hakodate National Hospital, Hokkaido, Japan
| | - Kei Hiraoka
- Department of Surgery, National Hospital Organization Hakodate National Hospital, Hokkaido, Japan
| | - Takanori Teshima
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, Japan.,Department of Hematology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Masaaki Murakami
- Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Team of Quantum immunology, Institute for Quantum Life Science, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan.,Division of Molecular Neuroimmunology, Department of Homeostatic Regulation, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan.,Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| |
Collapse
|
10
|
Kotaki R, Moriyama S, Takahashi Y. Humoral immunity for durable control of SARS-CoV-2 and its variants. Inflamm Regen 2023; 43:4. [PMID: 36631890 PMCID: PMC9834039 DOI: 10.1186/s41232-023-00255-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is ongoing because of the repeated emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants, highlighting the importance of developing vaccines for variants that may continue to emerge. In the present review, we discuss humoral immune responses against SARS-CoV-2 with a focus on the antibody breadth to the variants. Recent studies have revealed that the temporal maturation of humoral immunity improves the antibody potency and breadth to the variants after infection or vaccination. Repeated vaccination or infection further accelerates the expansion of the antibody breadth. Memory B cells play a central role in this phenomenon, as the reactivity of the B-cell antigen receptor (BCR) on memory B cells is a key determinant of the antibody potency and breadth recalled upon vaccination or infection. The evolution of memory B cells remarkably improves the reactivity of BCR to antigenically distinct Omicron variants, to which the host has never been exposed. Thus, the evolution of memory B cells toward the variants constitutes an immunological basis for the durable and broad control of SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Ryutaro Kotaki
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Saya Moriyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan.
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan.
| |
Collapse
|
11
|
Yang F, Zhu Z, Liu H, Cao W, Zhang W, Wei T, Zheng M, Zhang K, Tian H, Zeng Q, Cai X, Zheng H. Evaluation of Antibody Response in Sows after Vaccination with Senecavirus A Vaccine and the Effect of Maternal Antibody Transfer on Antibody Dynamics in Offspring. Vaccines (Basel) 2021; 9:vaccines9101066. [PMID: 34696174 PMCID: PMC8538203 DOI: 10.3390/vaccines9101066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/14/2021] [Accepted: 09/18/2021] [Indexed: 12/02/2022] Open
Abstract
Senecavirus A (SVA) is a newly porcine virus that has been detected in many countries since its first detection in pigs in Canada in 2007, and it remains endemic in many countries in Asia and America, which has become a substantial problem for the pig industry. Vaccination is a potentially effective strategy for the prevention and control of SVA infection. Our lab has developed a SVA vaccine candidate previously. In this study, the antibody response to the prepared vaccine in sows and their offspring was evaluated. Vaccination of sows with inactivated SVA vaccines during pregnancy elicited SVA-specific virus-neutralizing antibodies. Vaccination with a high dose of SVA vaccine followed a booster immunization contributed to a long-term duration of the persistence of maternally derived neutralizing antibodies (MDAs) in the milk of the sows (>14 days). In contrast, vaccination with a single low dose of SVA vaccine resulted in a short-term persistence of MDAs in the milk (2–7 days). The MDAs could be efficiently transferred from the sows to their offspring through the colostrum/milk but not the umbilical cord blood. The antibody titers and the duration of the persistence of MDAs in the offspring are highly associated with the antibody levels in the milk from the sows. Vaccination of sows with a booster dose of SVA vaccine resulted in a longer-lasting MDAs in their offspring (persisted for at least 90 days). However, vaccination with the single low dose of vaccine only brought about 42 days of MDAs persistence in their offspring. The effect of MDAs on active immunization with SVA vaccine in offspring was further evaluated, which showed that vaccination of the SVA vaccine in the presence of MDAs at the titer of ≈1:64 or less could overcome the MDAs’ interference and give rise to effective antibody response. This will help for establishing the optimal times and schedules for SVA vaccination in pigs.
Collapse
Affiliation(s)
- Fan Yang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (F.Y.); (Q.Z.)
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.Z.); (H.L.); (W.C.); (W.Z.); (T.W.); (M.Z.); (K.Z.); (H.T.)
| | - Zixiang Zhu
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.Z.); (H.L.); (W.C.); (W.Z.); (T.W.); (M.Z.); (K.Z.); (H.T.)
| | - Huanan Liu
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.Z.); (H.L.); (W.C.); (W.Z.); (T.W.); (M.Z.); (K.Z.); (H.T.)
| | - Weijun Cao
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.Z.); (H.L.); (W.C.); (W.Z.); (T.W.); (M.Z.); (K.Z.); (H.T.)
| | - Wei Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.Z.); (H.L.); (W.C.); (W.Z.); (T.W.); (M.Z.); (K.Z.); (H.T.)
| | - Ting Wei
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.Z.); (H.L.); (W.C.); (W.Z.); (T.W.); (M.Z.); (K.Z.); (H.T.)
| | - Min Zheng
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.Z.); (H.L.); (W.C.); (W.Z.); (T.W.); (M.Z.); (K.Z.); (H.T.)
| | - Keshan Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.Z.); (H.L.); (W.C.); (W.Z.); (T.W.); (M.Z.); (K.Z.); (H.T.)
| | - Hong Tian
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.Z.); (H.L.); (W.C.); (W.Z.); (T.W.); (M.Z.); (K.Z.); (H.T.)
| | - Qiaoying Zeng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (F.Y.); (Q.Z.)
| | - Xuepeng Cai
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (F.Y.); (Q.Z.)
- Correspondence: (X.C.); (H.Z.)
| | - Haixue Zheng
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Diseases Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.Z.); (H.L.); (W.C.); (W.Z.); (T.W.); (M.Z.); (K.Z.); (H.T.)
- Correspondence: (X.C.); (H.Z.)
| |
Collapse
|
12
|
Marcano VC, Cardenas-Garcia S, Diel DG, Antoniassi da Silva LH, Gogal RM, Miller PJ, Brown CC, Butt SL, Goraichuk IV, Dimitrov KM, Taylor TL, Williams-Coplin D, Olivier TL, Stanton JB, Afonso CL. A Novel Recombinant Newcastle Disease Vaccine Improves Post- In Ovo Vaccination Survival with Sustained Protection against Virulent Challenge. Vaccines (Basel) 2021; 9:vaccines9090953. [PMID: 34579191 PMCID: PMC8472951 DOI: 10.3390/vaccines9090953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/10/2021] [Accepted: 08/19/2021] [Indexed: 01/23/2023] Open
Abstract
In ovo vaccination has been employed by the poultry industry for over 20 years to control numerous avian diseases. Unfortunately, in ovo live vaccines against Newcastle disease have significant limitations, including high embryo mortality and the inability to induce full protection during the first two weeks of life. In this study, a recombinant live attenuated Newcastle disease virus vaccine containing the antisense sequence of chicken interleukin 4 (IL-4), rZJ1*L-IL4R, was used. The rZJ1*L-IL4R vaccine was administered in ovo to naïve specific pathogen free embryonated chicken eggs (ECEs) and evaluated against a homologous challenge. Controls included a live attenuated recombinant genotype VII vaccine based on the virus ZJ1 (rZJ1*L) backbone, the LaSota vaccine and diluent alone. In the first of two experiments, ECEs were vaccinated at 18 days of embryonation (DOE) with either 104.5 or 103.5 50% embryo infectious dose (EID50/egg) and chickens were challenged at 21 days post-hatch (DPH). In the second experiment, 103.5 EID50/egg of each vaccine was administered at 19 DOE, and chickens were challenged at 14 DPH. Chickens vaccinated with 103.5 EID50/egg of rZJ1*L-IL4R had hatch rates comparable to the group that received diluent alone, whereas other groups had significantly lower hatch rates. All vaccinated chickens survived challenge without displaying clinical disease, had protective hemagglutination inhibition titers, and shed comparable levels of challenge virus. The recombinant rZJ1*L-IL4R vaccine yielded lower post-vaccination mortality rates compared with the other in ovo NDV live vaccine candidates as well as provided strong protection post-challenge.
Collapse
Affiliation(s)
- Valerie C. Marcano
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Rd., Athens, GA 30605, USA; (V.C.M.); (S.C.-G.); (D.G.D.); (L.H.A.d.S.); (P.J.M.); (S.L.B.); (I.V.G.); (K.M.D.); (T.L.T.); (D.W.-C.); (T.L.O.)
- Department of Veterinary Pathology, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (C.C.B.); (J.B.S.)
| | - Stivalis Cardenas-Garcia
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Rd., Athens, GA 30605, USA; (V.C.M.); (S.C.-G.); (D.G.D.); (L.H.A.d.S.); (P.J.M.); (S.L.B.); (I.V.G.); (K.M.D.); (T.L.T.); (D.W.-C.); (T.L.O.)
- Department of Veterinary Pathology, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (C.C.B.); (J.B.S.)
| | - Diego G. Diel
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Rd., Athens, GA 30605, USA; (V.C.M.); (S.C.-G.); (D.G.D.); (L.H.A.d.S.); (P.J.M.); (S.L.B.); (I.V.G.); (K.M.D.); (T.L.T.); (D.W.-C.); (T.L.O.)
| | - Luciana H. Antoniassi da Silva
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Rd., Athens, GA 30605, USA; (V.C.M.); (S.C.-G.); (D.G.D.); (L.H.A.d.S.); (P.J.M.); (S.L.B.); (I.V.G.); (K.M.D.); (T.L.T.); (D.W.-C.); (T.L.O.)
| | - Robert M. Gogal
- Department of Veterinary Biosciences & Diagnostic Imaging, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA;
| | - Patti J. Miller
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Rd., Athens, GA 30605, USA; (V.C.M.); (S.C.-G.); (D.G.D.); (L.H.A.d.S.); (P.J.M.); (S.L.B.); (I.V.G.); (K.M.D.); (T.L.T.); (D.W.-C.); (T.L.O.)
| | - Corrie C. Brown
- Department of Veterinary Pathology, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (C.C.B.); (J.B.S.)
| | - Salman Latif Butt
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Rd., Athens, GA 30605, USA; (V.C.M.); (S.C.-G.); (D.G.D.); (L.H.A.d.S.); (P.J.M.); (S.L.B.); (I.V.G.); (K.M.D.); (T.L.T.); (D.W.-C.); (T.L.O.)
- Department of Veterinary Pathology, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (C.C.B.); (J.B.S.)
- Department of Pathology, UAF Sub Campus TTS, University of Agriculture Faisalabad, Punjab 38000, Pakistan
| | - Iryna V. Goraichuk
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Rd., Athens, GA 30605, USA; (V.C.M.); (S.C.-G.); (D.G.D.); (L.H.A.d.S.); (P.J.M.); (S.L.B.); (I.V.G.); (K.M.D.); (T.L.T.); (D.W.-C.); (T.L.O.)
- National Scientific Center Institute of Experimental and Clinical Veterinary Medicine, 83 Pushkinska St., 61023 Kharkiv, Ukraine
| | - Kiril M. Dimitrov
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Rd., Athens, GA 30605, USA; (V.C.M.); (S.C.-G.); (D.G.D.); (L.H.A.d.S.); (P.J.M.); (S.L.B.); (I.V.G.); (K.M.D.); (T.L.T.); (D.W.-C.); (T.L.O.)
| | - Tonya L. Taylor
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Rd., Athens, GA 30605, USA; (V.C.M.); (S.C.-G.); (D.G.D.); (L.H.A.d.S.); (P.J.M.); (S.L.B.); (I.V.G.); (K.M.D.); (T.L.T.); (D.W.-C.); (T.L.O.)
| | - Dawn Williams-Coplin
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Rd., Athens, GA 30605, USA; (V.C.M.); (S.C.-G.); (D.G.D.); (L.H.A.d.S.); (P.J.M.); (S.L.B.); (I.V.G.); (K.M.D.); (T.L.T.); (D.W.-C.); (T.L.O.)
| | - Timothy L. Olivier
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Rd., Athens, GA 30605, USA; (V.C.M.); (S.C.-G.); (D.G.D.); (L.H.A.d.S.); (P.J.M.); (S.L.B.); (I.V.G.); (K.M.D.); (T.L.T.); (D.W.-C.); (T.L.O.)
| | - James B. Stanton
- Department of Veterinary Pathology, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (C.C.B.); (J.B.S.)
| | - Claudio L. Afonso
- Exotic and Emerging Avian Viral Diseases Research Unit, Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, USDA, 934 College Station Rd., Athens, GA 30605, USA; (V.C.M.); (S.C.-G.); (D.G.D.); (L.H.A.d.S.); (P.J.M.); (S.L.B.); (I.V.G.); (K.M.D.); (T.L.T.); (D.W.-C.); (T.L.O.)
- Correspondence:
| |
Collapse
|
13
|
Otero CE, Langel SN, Blasi M, Permar SR. Maternal antibody interference contributes to reduced rotavirus vaccine efficacy in developing countries. PLoS Pathog 2020; 16:e1009010. [PMID: 33211756 PMCID: PMC7676686 DOI: 10.1371/journal.ppat.1009010] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Rotavirus (RV) vaccine efficacy is significantly reduced in lower- and middle-income countries (LMICs) compared to high-income countries. This review summarizes current research into the mechanisms behind this phenomenon, with a particular focus on the evidence that maternal antibody (matAb) interference is a contributing factor to this disparity. All RV vaccines currently in use are orally administered, live-attenuated virus vaccines that replicate in the infant gut, which leaves their efficacy potentially impacted by both placentally transferred immunoglobulin G (IgG) and mucosal IgA Abs conferred via breast milk. Observational studies of cohorts in LMICs demonstrated an inverse correlation between matAb titers, both in serum and breast milk, and infant responses to RV vaccination. However, a causal link between maternal humoral immunity and reduced RV vaccine efficacy in infants has yet to be definitively established, partially due to limitations in current animal models of RV disease. The characteristics of Abs mediating interference and the mechanism(s) involved have yet to be determined, and these may differ from mechanisms of matAb interference for parenterally administered vaccines due to the contribution of mucosal immunity conferred via breast milk. Increased vaccine doses and later age of vaccine administration have been strategies applied to overcome matAb interference, but these approaches are difficult to safely implement in the setting of RV vaccination in LMICs. Ultimately, the development of relevant animal models of matAb interference is needed to determine what alternative approaches or vaccine designs can safely and effectively overcome matAb interference of infant RV vaccination.
Collapse
Affiliation(s)
- Claire E. Otero
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pathology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Stephanie N. Langel
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Maria Blasi
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pathology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
14
|
Antibody Feedback Limits the Expansion of B Cell Responses to Malaria Vaccination but Drives Diversification of the Humoral Response. Cell Host Microbe 2020; 28:572-585.e7. [PMID: 32697938 DOI: 10.1016/j.chom.2020.07.001] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 06/02/2020] [Accepted: 06/30/2020] [Indexed: 12/20/2022]
Abstract
Generating sufficient antibody to block infection is a key challenge for vaccines against malaria. Here, we show that antibody titers to a key target, the repeat region of the Plasmodium falciparum circumsporozoite protein (PfCSP), plateaued after two immunizations in a clinical trial of the radiation-attenuated sporozoite vaccine. To understand the mechanisms limiting vaccine responsiveness, we developed immunoglobulin (Ig)-knockin mice with elevated numbers of PfCSP-binding B cells. We determined that recall responses were inhibited by antibody feedback, potentially via epitope masking of the immunodominant PfCSP repeat region. Importantly, the amount of antibody that prevents boosting is below the amount of antibody required for protection. Finally, while antibody feedback limited responses to the PfCSP repeat region in vaccinated volunteers, potentially protective subdominant responses to PfCSP C-terminal regions expanded with subsequent boosts. These data suggest that antibody feedback drives the diversification of immune responses and that vaccination for malaria will require targeting multiple antigens.
Collapse
|
15
|
Xu H, Heyman B. IgG-mediated suppression of antibody responses: Hiding or snatching epitopes? Scand J Immunol 2020; 92:e12921. [PMID: 32594540 DOI: 10.1111/sji.12921] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/08/2020] [Accepted: 06/21/2020] [Indexed: 01/03/2023]
Abstract
Antibodies forming a complex with antigen in vivo can dramatically change the antibody response to this antigen. In some situations, the response will be a 100-fold stronger than in animals immunized with antigen alone, and in other situations, the response will be completely suppressed. IgG is known to suppress the antibody response, for example to erythrocytes, and this is used clinically in Rhesus prophylaxis. The mechanism behind IgG-mediated immune suppression is still not understood. Here, we will review studies performed in experimental animal models and discuss the various hypotheses put forward to explain the profound suppressive effect of IgG. We conclude that an exclusive role for negative regulation of B cells through FcγRIIB, increased clearance of erythrocytes from the circulation or complement-mediated lysis is unlikely. Epitope masking, where IgG hides the epitope from B cells, or trogocytosis, where IgG removes the epitope from the erythrocyte, is compatible with many observations. These two mechanisms are not mutually exclusive. Moreover, it cannot be ruled out that clearance, in combination with other mechanisms, plays a role.
Collapse
Affiliation(s)
- Hui Xu
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Birgitta Heyman
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
16
|
Shinde P, Howie HL, Stegmann TC, Hay AM, Waterman HR, Szittner Z, Bentlage AEH, Kapp L, Lissenberg-Thunnissen SN, Dekkers G, Schasfoort RBM, Ratcliffe SJ, Smolkin ME, Vidarsson G, van der Schoot CE, Hudson KE, Zimring JC. IgG Subclass Determines Suppression Versus Enhancement of Humoral Alloimmunity to Kell RBC Antigens in Mice. Front Immunol 2020; 11:1516. [PMID: 32765523 PMCID: PMC7378678 DOI: 10.3389/fimmu.2020.01516] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/09/2020] [Indexed: 02/03/2023] Open
Abstract
It has long been appreciated that immunoglobulins are not just the effector endpoint of humoral immunity, but rather have a complex role in regulating antibody responses themselves. Donor derived anti-RhD IgG has been used for over 50 years as an immunoprophylactic to prevent maternal alloimmunization to RhD. Although anti-RhD has dramatically decreased rates of hemolytic disease of the fetus and newborn (for the RhD alloantigen), anti-RhD also fails in some cases, and can even paradoxically enhance immune responses in some circumstances. Attempts to generate a monoclonal anti-RhD have largely failed, with some monoclonals suppressing less than donor derived anti-RhD and others enhancing immunity. These difficulties likely result, in part, because the mechanism of anti-RhD remains unclear. However, substantial evidence exists to reject the common explanations of simple clearance of RhD + RBCs or masking of antigen. Donor derived anti-RhD is a mixture of 4 different IgG subtypes. To the best of our knowledge an analysis of the role different IgG subtypes play in immunoregulation has not been carried out; and, only IgG1 and IgG3 have been tested as monoclonals. Multiple attempts to elicit alloimmune responses to human RhD epitopes in mice have failed. To circumvent this limitation, we utilize a tractable animal model of RBC alloimmunization using the human Kell glycoprotein as an antigen to test the effect of IgG subtype on immunoregulation by antibodies to RBC alloantigens. We report that the ability of an anti-RBC IgG to enhance, suppress (at the level of IgM responses), or have no effect is a function of the IgG subclass in this model system.
Collapse
Affiliation(s)
- Paurvi Shinde
- Bloodworks Northwest Research Institute, Seattle, WA, United States
| | - Heather L. Howie
- Department of Pathology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Tamara C. Stegmann
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Ariel M. Hay
- Department of Pathology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | | | - Zoltan Szittner
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Arthur E. H. Bentlage
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Linda Kapp
- Bloodworks Northwest Research Institute, Seattle, WA, United States
| | - Suzanne N. Lissenberg-Thunnissen
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Gillian Dekkers
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | | | - Sarah J. Ratcliffe
- Department of Pathology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Mark E. Smolkin
- Department of Pathology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Gestur Vidarsson
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - C. Ellen van der Schoot
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Krystalyn E. Hudson
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States
| | - James C. Zimring
- Bloodworks Northwest Research Institute, Seattle, WA, United States
- Department of Pathology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
17
|
Anania JC, Westin A, Heyman B. IgG Suppresses Antibody Responses to Sheep Red Blood Cells in Double Knock-Out Mice Lacking Complement Factor C3 and Activating Fcγ-Receptors. Front Immunol 2020; 11:1404. [PMID: 32733467 PMCID: PMC7360818 DOI: 10.3389/fimmu.2020.01404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022] Open
Abstract
Antigen-specific IgG antibodies, passively administered together with erythrocytes, prevent antibody responses against the erythrocytes. The mechanism behind the suppressive ability of IgG has been the subject of intensive studies, yet there is no consensus as to how it works. An important question is whether the Fc-region of IgG is required. Several laboratories have shown that IgG suppresses equally well in wildtype mice and mice lacking the inhibitory FcγIIB, activating FcγRs (FcγRI, III, and IV), or complement factor C3. These observations consistently suggest that IgG-mediated suppression does not rely on Fc-mediated antibody functions. However, it was recently shown that anti-KEL sera failed to suppress antibody responses to KEL-expressing transgenic mouse erythrocytes in double knock-out mice lacking both activating FcγRs and C3. Yet, in the same study, antibody-mediated suppression worked well in each single knock-out strain. This unexpected observation suggested Fc-dependence of IgG-mediated suppression and prompted us to investigate the issue in the classical experimental model using sheep red blood cells (SRBC) as antigen. SRBC alone or IgG anti-SRBC together with SRBC was administered to wildtype and double knock-out mice lacking C3 and activating FcγRs. IgG efficiently suppressed the IgM and IgG anti-SRBC responses in both mouse strains, thus supporting previous observations that suppression in this model is Fc-independent.
Collapse
Affiliation(s)
- Jessica C Anania
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Annika Westin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Birgitta Heyman
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
18
|
Knight M, Changrob S, Li L, Wilson PC. Imprinting, immunodominance, and other impediments to generating broad influenza immunity. Immunol Rev 2020; 296:191-204. [PMID: 32666572 DOI: 10.1111/imr.12900] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022]
Abstract
Natural influenza virus infections and seasonal vaccinations often do not confer broadly neutralizing immunity across diverse influenza strains. In addition, the virus is capable of rapid antigenic drift in order to evade pre-existing immunity. The surface glycoproteins, hemagglutinin, and neuraminidase can easily mutate their immunodominant epitopes without impacting fitness. Skewing human antibody repertoires to target more conserved epitopes is thus an expanding area of research: Many groups are attempting to produce universal influenza vaccines that can protect across a wide variety of strains. Achieving this goal will require a detailed understanding of how infection history impacts humoral responses. It will also require the ability to manipulate or enhance B cell selection in order to expand clones that can recognize subdominant but protective epitopes. In this review, we will discuss what immune imprinting means to immunologists and describe efforts to overcome or silence imprinting in order to improve vaccination efficiency.
Collapse
Affiliation(s)
- Matthew Knight
- Department of Medicine, Section of Rheumatology, The Knapp Center for Lupus and Immunology Research, The University of Chicago, Chicago, IL, USA
| | - Siriruk Changrob
- Department of Medicine, Section of Rheumatology, The Knapp Center for Lupus and Immunology Research, The University of Chicago, Chicago, IL, USA
| | - Lei Li
- Department of Medicine, Section of Rheumatology, The Knapp Center for Lupus and Immunology Research, The University of Chicago, Chicago, IL, USA
| | - Patrick C Wilson
- Department of Medicine, Section of Rheumatology, The Knapp Center for Lupus and Immunology Research, The University of Chicago, Chicago, IL, USA
- Committee on Immunology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
19
|
Hu Z, Ni J, Cao Y, Liu X. Newcastle Disease Virus as a Vaccine Vector for 20 Years: A Focus on Maternally Derived Antibody Interference. Vaccines (Basel) 2020; 8:vaccines8020222. [PMID: 32422944 PMCID: PMC7349365 DOI: 10.3390/vaccines8020222] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/28/2020] [Accepted: 05/11/2020] [Indexed: 01/08/2023] Open
Abstract
It has been 20 years since Newcastle disease virus (NDV) was first used as a vector. The past two decades have witnessed remarkable progress in vaccine generation based on the NDV vector and optimization of the vector. Protective antigens of a variety of pathogens have been expressed in the NDV vector to generate novel vaccines for animals and humans, highlighting a great potential of NDV as a vaccine vector. More importantly, the research work also unveils a major problem restraining the NDV vector vaccines in poultry, i.e., the interference from maternally derived antibody (MDA). Although many efforts have been taken to overcome MDA interference, a lack of understanding of the mechanism of vaccination inhibition by MDA in poultry still hinders vaccine improvement. In this review, we outline the history of NDV as a vaccine vector by highlighting some milestones. The recent advances in the development of NDV-vectored vaccines or therapeutics for animals and humans are discussed. Particularly, we focus on the mechanisms and hypotheses of vaccination inhibition by MDA and the efforts to circumvent MDA interference with the NDV vector vaccines. Perspectives to fill the gap of understanding concerning the mechanism of MDA interference in poultry and to improve the NDV vector vaccines are also proposed.
Collapse
Affiliation(s)
- Zenglei Hu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Jie Ni
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Yongzhong Cao
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
| | - Xiufan Liu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
20
|
Gruber DR, Richards AL, Howie HL, Hay AM, Lebedev JN, Wang X, Zimring JC, Hudson KE. Passively transferred IgG enhances humoral immunity to a red blood cell alloantigen in mice. Blood Adv 2020; 4:1526-1537. [PMID: 32289162 PMCID: PMC7160277 DOI: 10.1182/bloodadvances.2019001299] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/10/2020] [Indexed: 12/16/2022] Open
Abstract
Antibodies are typically thought of as the endpoint of humoral immunity that occur as the result of an adaptive immune response. However, affinity-matured antibodies can be present at the initiation of a new immune response, most commonly because of passive administration as a medical therapy. The current paradigm is that immunoglobulin M (IgM), IgA, and IgE enhance subsequent humoral immunity. In contrast, IgG has a "dual effect" in which it enhances responses to soluble antigens but suppresses responses to antigens on red blood cells (RBCs) (eg, immunoprophylaxis with anti-RhD). Here, we report a system in which passive antibody to an RBC antigen promotes a robust cellular immune response leading to endogenous CD4+ T-cell activation, germinal center formation, antibody secretion, and immunological memory. The mechanism requires ligation of Fcγ receptors on a specific subset of dendritic cells that results in CD4+ T-cell activation and expansion. Moreover, antibodies cross-enhance responses to a third-party antigen, but only if it is expressed on the same RBC as the antigen recognized by the antibody. Importantly, these observations were IgG subtype specific. Thus, these findings demonstrate that antibodies to RBC alloantigens can enhance humoral immunity in an IgG subtype-specific fashion and provide mechanistic elucidation of the enhancing effects.
Collapse
Affiliation(s)
| | | | - Heather L Howie
- Department of Pathology and
- Carter Immunology Center, University of Virginia, Charlottesville, VA; and
| | | | | | | | - James C Zimring
- Department of Pathology and
- Carter Immunology Center, University of Virginia, Charlottesville, VA; and
| | - Krystalyn E Hudson
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
21
|
Arulraj T, Binder SC, Robert PA, Meyer-Hermann M. Synchronous Germinal Center Onset Impacts the Efficiency of Antibody Responses. Front Immunol 2019; 10:2116. [PMID: 31555300 PMCID: PMC6742702 DOI: 10.3389/fimmu.2019.02116] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 08/22/2019] [Indexed: 12/25/2022] Open
Abstract
The germinal center reaction is an important target for modulating antibody responses. Antibody production from germinal centers is regulated by a negative feedback mechanism termed antibody feedback. By imposing antibody feedback, germinal centers can interact and regulate the output of other germinal centers. Using an agent-based model of the germinal center reaction, we studied the impact of antibody feedback on kinetics and efficiency of a germinal center. Our simulations predict that high feedback of antibodies from germinal centers reduces the production of plasma cells and subsequently the efficiency of the germinal center reaction by promoting earlier termination. Affinity maturation is only weakly improved by increased antibody feedback and ultimately interrupted because of premature termination of the reaction. The model predicts that the asynchronous onset and changes in number of germinal centers could alter the efficiency of antibody response due to changes in feedback by soluble antibodies. Consequently, late initialized germinal centers have a compromised output due to higher antibody feedback from the germinal centers formed earlier. The results demonstrate potential effects of germinal center intercommunication and highlight the importance of understanding germinal center interactions for optimizing the antibody response, in particular, in the elderly and in the context of vaccination.
Collapse
Affiliation(s)
- Theinmozhi Arulraj
- Department of Systems Immunology, Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Sebastian C Binder
- Department of Systems Immunology, Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Centre for Individualized Infection Medicine (CIIM), Hanover, Germany
| | - Philippe A Robert
- Department of Systems Immunology, Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology, Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Centre for Individualized Infection Medicine (CIIM), Hanover, Germany.,Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
22
|
Abstract
Specific IgM, administered together with the antigen it recognizes, enhances primary antibody responses, formation of germinal centers, and priming for secondary antibody responses. The response to all epitopes on the antigen to which IgM binds is usually enhanced. IgM preferentially enhances responses to large antigens such as erythrocytes, malaria parasites, and keyhole limpet hemocyanine. In order for an effect to be seen, antigens must be administered in suboptimal concentrations and in close temporal relationship to the IgM. Enhancement is dependent on the ability of IgM to activate complement, but the lytic pathway is not required. Enhancement does not take place in mice lacking complement receptors 1 and 2 (CR1/2) suggesting that the role of IgM is to generate C3 split products, i.e., the ligands for CR1/2. In mice, these receptors are expressed on follicular dendritic cells (FDCs) and B cells. Optimal IgM-mediated enhancement requires that both cell types express CR1/2, but intermediate enhancement is seen when only FDCs express the receptors and low enhancement when only B cells express them. These observations imply that IgM-mediated enhancement works through several, non-mutually exclusive, pathways. Marginal zone B cells can transport IgM-antigen-complement complexes, bound to CR1/2, from the marginal zone and deposit them onto FDCs. In addition, co-crosslinking of the BCR and the CR2/CD19/CD81 co-receptor complex may enhance signaling to specific B cells, a mechanism likely to be involved in induction of early extrafollicular antibody responses.
Collapse
Affiliation(s)
- Anna Sörman
- Department of Medical Biochemistry and Microbiology, Uppsala University, BMC, Box 582, SE 751 23, Uppsala, Sweden
| | - Birgitta Heyman
- Department of Medical Biochemistry and Microbiology, Uppsala University, BMC, Box 582, SE 751 23, Uppsala, Sweden.
| |
Collapse
|
23
|
Xu H, Zhang L, Heyman B. IgG-mediated immune suppression in mice is epitope specific except during high epitope density conditions. Sci Rep 2018; 8:15292. [PMID: 30327481 PMCID: PMC6191431 DOI: 10.1038/s41598-018-33087-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 09/12/2018] [Indexed: 01/12/2023] Open
Abstract
Specific IgG antibodies, passively administered together with erythrocytes, suppress antibody responses against the erythrocytes. Although used to prevent alloimmunization in Rhesus (Rh)D-negative women carrying RhD-positive fetuses, the mechanism behind is not understood. In mice, IgG suppresses efficiently in the absence of Fcγ-receptors and complement, suggesting an Fc-independent mechanism. In line with this, suppression is frequently restricted to the epitopes to which IgG binds. However, suppression of responses against epitopes not recognized by IgG has also been observed thus arguing against Fc-independence. Here, we explored the possibility that non-epitope specific suppression can be explained by steric hindrance when the suppressive IgG binds to an epitope present at high density. Mice were transfused with IgG anti-4-hydroxy-3-nitrophenylacetyl (NP) together with NP-conjugated sheep red blood cells (SRBC) with high, intermediate, or low NP-density. Antibody titers and the number of single antibody-forming cells were determined. As a rule, IgG suppressed NP- but not SRBC-specific responses (epitope specific suppression). However, there was one exception: suppression of both IgM anti-SRBC and IgM anti-NP responses occurred when high density SRBC-NP was administered (non-epitope specific suppression). These findings answer a longstanding question in antibody feedback regulation and are compatible with the hypothesis that epitope masking explains IgG-mediated immune suppression.
Collapse
Affiliation(s)
- Hui Xu
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Lu Zhang
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Birgitta Heyman
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
24
|
Pashov A, Hernandez Puente CV, Ibrahim SM, Monzavi-Karbassi B, Makhoul I, Kieber-Emmons T. Thinking Cancer. Monoclon Antib Immunodiagn Immunother 2018; 37:117-125. [PMID: 29939836 DOI: 10.1089/mab.2018.0014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Evolutionary theories are necessarily invoked for understanding cancer development at the level of species, at the level of cells and tissues, and for developing effective therapies. It is crucial to view cancer in a Darwinian light, where the differential survival of individual cells is based on heritable variations. In the process of this somatic evolution, multicellularity controls are overridden by cancer cells, which become increasingly autonomous. Ecological epigenetics also helps understand how rogue cells that have basically the same DNA as their normal cell counterpart overcome the tissue homeostasis. As we struggle to wrap our minds around the complexity of these phenomena, we apply often times anthropomorphic terms, such as subversion, hijacking, or hacking, to describe especially the most complex among them-the interaction of tumors with the immune system. In this commentary we highlight examples of the anthropomorphic thinking of cancer and try to put into context the relative meaning of terms and the mechanisms that are oftentimes invoked to justify those terms.
Collapse
Affiliation(s)
- Anastas Pashov
- 1 Stephan Angelov Institute of Microbiology , Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | | | - Behjatolah Monzavi-Karbassi
- 3 Department of Pathology, University of Arkansas for Medical Sciences , Little Rock, Arkansas
- 4 Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| | - Issam Makhoul
- 4 Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences , Little Rock, Arkansas
- 5 Department of Medicine, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| | - Thomas Kieber-Emmons
- 3 Department of Pathology, University of Arkansas for Medical Sciences , Little Rock, Arkansas
- 4 Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences , Little Rock, Arkansas
| |
Collapse
|
25
|
Zwiers C, Koelewijn JM, Vermij L, van Sambeeck J, Oepkes D, de Haas M, van der Schoot CE. ABO incompatibility and RhIG immunoprophylaxis protect against non-D alloimmunization by pregnancy. Transfusion 2018; 58:1611-1617. [PMID: 29624682 DOI: 10.1111/trf.14606] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/19/2018] [Accepted: 02/21/2018] [Indexed: 01/19/2023]
Abstract
BACKGROUND Hemolytic disease of the fetus and newborn (HDFN) is caused by maternal antibodies against fetal red blood cell antigens, most often anti-D, -K, or -c. ABO incompatibility between mother and child and anti-D immunoprophylaxis (RhIG) are known to reduce the risk of D immunization and subsequent HDFN. However, no immunoprophylaxis has been developed to prevent non-D immunizations. STUDY DESIGN AND METHODS We evaluated whether ABO incompatibility has a preventive effect on formation of non-D alloantibodies, by performing a case-control study including pregnant women with newly detected non-D antibodies, identified within a nationwide data set, immunized during their first pregnancy and/or delivery. Subsequently, we assessed a possible protective effect of RhIG in a subgroup with non-Rh antibodies only. The proportions of previous ABO incompatibility and of RhIG administrations of these women were compared to the known rate of 19.4% ABO incompatibility and 9.9% RhIG administrations (D- women carrying a D+ child) in the general population of pregnant women. RESULTS A total of 11.9% of the 232 included immunized women had a possible ABO incompatibility in their first pregnancy (vs. expected 19.4%; 95% confidence interval [CI], 7.3-18.8; p = 0.036). Furthermore, 1.0% women with non-Rh antibodies were D-, delivered a D+ child, and had therefore received RhIG, whereas 9.9% was expected (95% CI, 0.18-5.50; p = 0.003). CONCLUSION We found that ABO incompatibility and RhIG reduce the risks not only for D, but also for non-Rh immunizations, suggesting that antibody-mediated immune suppression in this condition is not antigen specific.
Collapse
Affiliation(s)
- Carolien Zwiers
- Department of Obstetrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Joke M Koelewijn
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam.,Sanquin Diagnostic Services, Amsterdam, the Netherlands
| | - Lisa Vermij
- Department of Obstetrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Joost van Sambeeck
- Department of Transfusion Technology Assessment, Sanquin Research, Amsterdam.,Centre for Healthcare Operations Improvement & Research, University of Twente, Enschede, the Netherlands
| | - Dick Oepkes
- Department of Obstetrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Masja de Haas
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands.,Sanquin Diagnostic Services, Amsterdam, the Netherlands.,Center for Clinical Transfusion Research, Sanquin Research, Leiden, the Netherlands
| | - C Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam
| |
Collapse
|
26
|
Cruz-Leal Y, Marjoram D, Lazarus AH. Erythrocyte Saturation with IgG Is Required for Inducing Antibody-Mediated Immune Suppression and Impacts Both Erythrocyte Clearance and Antigen-Modulation Mechanisms. THE JOURNAL OF IMMUNOLOGY 2018; 200:1295-1305. [PMID: 29358275 DOI: 10.4049/jimmunol.1700874] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 12/04/2017] [Indexed: 12/21/2022]
Abstract
Anti-D prevents hemolytic disease of the fetus and newborn, and this mechanism has been referred to as Ab-mediated immune suppression (AMIS). Anti-D, as well as other polyclonal AMIS-inducing Abs, most often induce both epitope masking and erythrocyte clearance mechanisms. We have previously observed that some Abs that successfully induce AMIS effects could be split into those that mediate epitope masking versus those that induce erythrocyte clearance, allowing the ability to analyze these mechanisms separately. In addition, AMIS-inducing activity has recently been shown to induce Ag modulation (Ag loss from the erythrocyte surface). To assess these mechanisms, we immunized mice with transgenic murine RBCs expressing a single Ag protein comprising a recombinant Ag composed of hen egg lysozyme, OVA sequences comprising aa 251-349, and the human Duffy transmembrane protein (HOD-Ag) with serial doses of polyclonal anti-OVA IgG as the AMIS-inducing Ab. The anti-OVA Ab induced AMIS in the absence of apparent epitope masking. AMIS occurred only when the erythrocytes appeared saturated with IgG. This Ab was capable of inducing HOD-RBC clearance, as well as loss of the OVA epitope at doses of Ab that caused AMIS effects. HOD-RBCs also lost reactivity with Abs specific for the hen egg lysozyme and Duffy portions of the Ag consistent with the initiation of Ag modulation and/or trogocytosis mechanisms. These data support the concept that an AMIS-inducing Ab that does not cause epitope masking can induce AMIS effects in a manner consistent with RBC clearance and/or Ag modulation.
Collapse
Affiliation(s)
- Yoelys Cruz-Leal
- Department of Laboratory Medicine and the Keenan Research Centre for Biomedical Science in the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada.,Centre for Innovation, Canadian Blood Services, Ottawa, Ontario K1G 4J5, Canada
| | - Danielle Marjoram
- Department of Laboratory Medicine and the Keenan Research Centre for Biomedical Science in the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
| | - Alan H Lazarus
- Department of Laboratory Medicine and the Keenan Research Centre for Biomedical Science in the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada; .,Centre for Innovation, Canadian Blood Services, Ottawa, Ontario K1G 4J5, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario M5G 2C4, Canada; and.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
27
|
|
28
|
Bergström JJE, Heyman B. Mice Immunized with IgG Anti-Sheep Red Blood Cells (SRBC) Together With SRBC Have a Suppressed Anti-SRBC Antibody Response but Generate Germinal Centers and Anti-IgG Antibodies in Response to the Passively Administered IgG. Front Immunol 2017; 8:911. [PMID: 28824636 PMCID: PMC5539184 DOI: 10.3389/fimmu.2017.00911] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/17/2017] [Indexed: 12/29/2022] Open
Abstract
Antigen-specific IgG antibodies, passively administered together with large particulate antigens such as erythrocytes, can completely suppress the antigen-specific antibody response. The mechanism behind has been elusive. Herein, we made the surprising observation that mice immunized with IgG anti-sheep red blood cells (SRBC) and SRBC, in spite of a severely suppressed anti-SRBC response, have a strong germinal center (GC) response. This occurred regardless of whether the passively administered IgG was of the same allotype as that of the recipient or not. Six days after immunization, the GC size and the number of GC B cells were higher in mice immunized with SRBC alone than in mice immunized with IgG and SRBC, but at the other time points these parameters were similar. GCs in the IgG-groups had a slight shift toward dark zone B cells 6 days after immunization and toward light zone B cells 10 days after immunization. The proportions of T follicular helper cells (TFH) and T follicular regulatory cells (TFR) were similar in the two groups. Interestingly, mice immunized with allogeneic IgG anti-SRBC together with SRBC mounted a vigorous antibody response against the passively administered suppressive IgG. Thus, although their anti-SRBC response was almost completely suppressed, an antibody response against allogeneic, and probably also syngeneic, IgG developed. This most likely explains the development of GCs in the absence of an anti-SRBC antibody response.
Collapse
Affiliation(s)
- Joakim J E Bergström
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Birgitta Heyman
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
29
|
Bergström JJE, Xu H, Heyman B. Epitope-Specific Suppression of IgG Responses by Passively Administered Specific IgG: Evidence of Epitope Masking. Front Immunol 2017; 8:238. [PMID: 28321225 PMCID: PMC5337509 DOI: 10.3389/fimmu.2017.00238] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 02/20/2017] [Indexed: 01/03/2023] Open
Abstract
Specific IgG, passively administered together with particulate antigen, can completely prevent induction of antibody responses to this antigen. The ability of IgG to suppress antibody responses to sheep red blood cells (SRBCs) is intact in mice lacking FcγRs, complement factor 1q, C3, or complement receptors 1 and 2, suggesting that Fc-dependent effector functions are not involved. Two of the most widely discussed explanations for the suppressive effect are increased clearance of IgG–antigen complexes and/or that IgG “hides” the antigen from recognition by specific B cells, so-called epitope masking. The majority of data on how IgG induces suppression was obtained through studies of the effects on IgM-secreting single spleen cells during the first week after immunization. Here, we show that IgG also suppresses antigen-specific extrafollicular antibody-secreting cells, germinal center B-cells, long-lived plasma cells, long-term IgG responses, and induction of memory antibody responses. IgG anti-SRBC reduced the amount of SRBC in the spleens of wild-type, but not of FcγR-deficient mice. However, no correlation between suppression and the amount of SRBC in the spleen was observed, suggesting that increased clearance does not explain IgG-mediated suppression. Instead, we found compelling evidence for epitope masking because IgG anti-NP administered with NP-SRBC suppressed the IgG anti-NP, but not the IgG anti-SRBC response. Vice versa, IgG anti-SRBC administered with NP-SRBC, suppressed only the IgG anti-SRBC response. In conclusion, passively transferred IgG suppressed all measured parameters of an antigen-specific antibody/B cell response and an important mechanism of action is likely to be epitope masking.
Collapse
Affiliation(s)
- Joakim J E Bergström
- Department of Medical Biochemistry and Microbiology, Uppsala University , Uppsala , Sweden
| | - Hui Xu
- Department of Medical Biochemistry and Microbiology, Uppsala University , Uppsala , Sweden
| | - Birgitta Heyman
- Department of Medical Biochemistry and Microbiology, Uppsala University , Uppsala , Sweden
| |
Collapse
|
30
|
Marjoram D, Cruz-Leal Y, Bernardo L, Lazarus AH. A role for red cell clearance in antibody-mediated inhibition of erythrocyte alloimmunization? ACTA ACUST UNITED AC 2016. [DOI: 10.1111/voxs.12320] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- D. Marjoram
- Department of Laboratory Medicine; Keenan Research Centre; Li Ka Shing Knowledge Institute; St. Michael's Hospital; Toronto ON Canada
- Department of Medicine and Laboratory Medicine and Pathobiology; University of Toronto; Toronto ON Canada
| | - Y. Cruz-Leal
- Department of Laboratory Medicine; Keenan Research Centre; Li Ka Shing Knowledge Institute; St. Michael's Hospital; Toronto ON Canada
| | - L. Bernardo
- Department of Laboratory Medicine; Keenan Research Centre; Li Ka Shing Knowledge Institute; St. Michael's Hospital; Toronto ON Canada
- The Canadian Blood Services; Toronto ON Canada
| | - A. H. Lazarus
- Department of Laboratory Medicine; Keenan Research Centre; Li Ka Shing Knowledge Institute; St. Michael's Hospital; Toronto ON Canada
- Department of Medicine and Laboratory Medicine and Pathobiology; University of Toronto; Toronto ON Canada
- The Canadian Blood Services; Toronto ON Canada
| |
Collapse
|
31
|
Victor JR. Allergen-specific IgG as a mediator of allergy inhibition: Lessons from mother to child. Hum Vaccin Immunother 2016; 13:507-513. [PMID: 27808600 PMCID: PMC5360138 DOI: 10.1080/21645515.2016.1244592] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Allergen-specific IgG produced by immune mothers is associated with less predisposition to allergy development in their children. This finding has been described by several groups over the last few decades, but the mechanisms by which maternal IgG can inhibit allergy development are still not fully understood. With the purpose of summarizing past investigations, we review the literature on murine models of maternal immunization with allergens and on immune regulation in humans after passive therapy with purified IgG. Based on our review, a new hypothesis about these mechanisms is presented, which may provide a foundation for the future development of therapies to inhibit allergy development.
Collapse
Affiliation(s)
- Jefferson Russo Victor
- a Laboratory of Medical Investigation LIM-56, Division of Clinical Dermatology , Medical School, University of Sao Paulo, Sao Paulo, Brazil; Division of Pathology, Medical School, University of Sao Paulo , Sao Paulo , Brazil
| |
Collapse
|
32
|
Chernyshov VP, Dons’koi BV, Sudoma IO, Goncharova YO. Multiple immune deviations predictive for IVF failure as possible markers for IVIG therapy. Immunol Lett 2016; 176:44-50. [DOI: 10.1016/j.imlet.2015.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/21/2015] [Accepted: 12/29/2015] [Indexed: 11/25/2022]
|
33
|
Triolo G, Ferrante A, Accardo-Palumbo A, Ciccia F, Cadelo M, Castelli A, Perino A, Licata G. IVIG in APS pregnancy. Lupus 2016; 13:731-5. [PMID: 15485113 DOI: 10.1191/0961203304lu2011oa] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
For more than two decades, the intravenous administration of high doses of IgG pooled from the plasma of healthy donors (immune globulin therapy, also known as ‘IVIG’) has benefited patients with a variety of autoimmune disorders. A potential therapeutic role of IVIG in the prevention of thrombosis and of miscarriages in antiphospholipid syndrome (APS) has been postulated. Multicenter randomized controlled trials attempted to define the role of IVIG in preventing pregnancy complications in APS indicate that simple anticoagulation could not be completely satisfactory, and certain patient subgroups might take advantage of IVIG therapy alone or in combination with heparin.
Collapse
Affiliation(s)
- G Triolo
- Sezione di Reumatologia, Dipartimento Biomedico di Medicina Interna e Specialistica, University of Palermo, Palermo, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Antibody-mediated immune suppression is improved when blends of anti-RBC monoclonal antibodies are used in mice. Blood 2016; 128:1076-80. [PMID: 27330002 DOI: 10.1182/blood-2016-01-692178] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 06/09/2016] [Indexed: 01/18/2023] Open
Abstract
Although the prevention of hemolytic disease of the fetus and newborn is highly effective using polyclonal anti-D, a recombinant alternative is long overdue. Unfortunately, anti-D monoclonal antibodies have been, at best, disappointing. To determine the primary attribute defining an optimal antibody, we assessed suppression of murine red blood cell (RBC) immunization by single-monoclonal antibodies vs defined blends of subtype-matched antibodies. Allogeneic RBCs expressing the HOD antigen (hen egg lysozyme [HEL]-ovalbumin-human transmembrane Duffy(b)) were transfused into naïve mice alone or together with selected combinations of HEL-specific antibodies, and the resulting suppressive effect was assessed by evaluating the antibody response. Polyclonal HEL antibodies dramatically inhibited the antibody response to the HOD antigen, whereas single-monoclonal HEL antibodies were less effective despite the use of saturating doses. A blend of monoclonal HEL-specific antibodies reactive with different HEL epitopes significantly increased the suppressive effect, whereas a blend of monoclonal antibodies that block each other's binding to the HEL protein did not increase suppression. In conclusion, these data show that polyclonal antibodies are superior to monoclonal antibodies at suppressing the immune response to the HOD cells, a feature that can be completely recapitulated using monoclonal antibodies to different epitopes.
Collapse
|
35
|
Fc Receptors and Fc Receptor-Like Molecules within the Immunoreceptor Family. ENCYCLOPEDIA OF IMMUNOBIOLOGY 2016. [PMCID: PMC7152311 DOI: 10.1016/b978-0-12-374279-7.02017-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Receptors for the Fc portion of immunoglobulins (FcRs) account for most cell-mediated biological activities of antibodies. The majority of FcRs are encoded by a set of genes, clustered in the fcr locus, on chromosome 1 in humans and on chromosome 1 and 3 in mice. Eight (in humans) and six (in mice) new genes were found, intermixed with FcR genes in corresponding fcr loci, which encode FcR-like molecules (FcRLs). FcRs and FcRLs are genetically, phylogenetically, structurally, and functionally related. FcRs and FcRLs, however, markedly differ by their ligands, their tissue distribution, and, therefore, by the biological functions they control. A systematic comparison of their biological properties leads to the conclusion that FcRLs are not like FcRs. They altogether form a single family within the immunoreceptor family, whose members fulfill distinct but complementary roles in immunity by differentially controlling innate and adaptive responses.
Collapse
|
36
|
Bergström JJE, Heyman B. IgG Suppresses Antibody Responses in Mice Lacking C1q, C3, Complement Receptors 1 and 2, or IgG Fc-Receptors. PLoS One 2015; 10:e0143841. [PMID: 26619292 PMCID: PMC4664261 DOI: 10.1371/journal.pone.0143841] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 11/10/2015] [Indexed: 11/21/2022] Open
Abstract
Antigen-specific IgG antibodies, passively administered to mice or humans together with large particulate antigens like erythrocytes, can completely suppress the antibody response against the antigen. This is used clinically in Rhesus prophylaxis, where administration of IgG anti-RhD prevents RhD-negative women from becoming immunized against RhD-positive fetal erythrocytes aquired transplacentally. The mechanisms by which IgG suppresses antibody responses are poorly understood. We have here addressed whether complement or Fc-receptors for IgG (FcγRs) are required for IgG-mediated suppression. IgG, specific for sheep red blood cells (SRBC), was administered to mice together with SRBC and the antibody responses analyzed. IgG was able to suppress early IgM- as well as longterm IgG-responses in wildtype mice equally well as in mice lacking FcγRIIB (FcγRIIB knockout mice) or FcγRI, III, and IV (FcRγ knockout mice). Moreover, IgG was able to suppress early IgM responses equally well in mice lacking C1q (C1qA knockout mice), C3 (C3 knockout mice), or complement receptors 1 and 2 (Cr2 knockout mice) as in wildtype mice. Owing to the previously described severely impaired IgG responses in the complement deficient mice, it was difficult to assess whether passively administered IgG further decreased their IgG response. In conclusion, Fc-receptor binding or complement-activation by IgG does not seem to be required for its ability to suppress antibody responses to xenogeneic erythrocytes.
Collapse
Affiliation(s)
- Joakim J. E. Bergström
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Birgitta Heyman
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
37
|
Edwards KM. Maternal antibodies and infant immune responses to vaccines. Vaccine 2015; 33:6469-72. [PMID: 26256526 DOI: 10.1016/j.vaccine.2015.07.085] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/15/2015] [Accepted: 07/17/2015] [Indexed: 11/17/2022]
Abstract
Infants are born with immature immune systems, making it difficult for them to effectively respond to the infectious pathogens encountered shortly after birth. Maternal antibody is actively transported across the placenta and serves to provide protection to the newborn during the first weeks to months of life. However, maternal antibody has been shown repeatedly to inhibit the immune responses of young children to vaccines. The mechanisms for this inhibition are presented and the impact on ultimate immune responses is discussed.
Collapse
Affiliation(s)
- Kathryn M Edwards
- Sarah H. Sell and Cornelius Vanderbilt Chair in Pediatrics, Vanderbilt Vaccine Research Program, Department of Pediatrics, Vanderbilt University, 21st Avenue South, S 2323 MCN, Nashville, TN 37232, United States.
| |
Collapse
|
38
|
Bernardo L, Yu H, Amash A, Zimring JC, Lazarus AH. IgG-Mediated Immune Suppression to Erythrocytes by Polyclonal Antibodies Can Occur in the Absence of Activating or Inhibitory Fcγ Receptors in a Full Mouse Model. THE JOURNAL OF IMMUNOLOGY 2015; 195:2224-30. [PMID: 26188060 DOI: 10.4049/jimmunol.1500790] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 06/22/2015] [Indexed: 01/01/2023]
Abstract
Polyclonal anti-D has been used to prevent RhD-negative mothers from becoming immunized against RhD positive fetal erythrocytes, and this mechanism has been referred as Ab or IgG-mediated immune suppression (AMIS). Although anti-D has been highly successful, the inhibitory mechanisms remain poorly understood. Two major theories behind AMIS involve the binding of IgG to activating or inhibitory FcγR, which can induce either erythrocyte clearance or immune inhibition, respectively. In this work, we explored the absolute role of activating and inhibitory FcγR in the AMIS mechanism using the HOD mouse model of RBC immunization. HOD mice contain a RBC-specific recombinant protein composed of hen egg lysozyme (HEL), OVA and human transmembrane Duffy Ag, and erythrocytes from HOD mice can stimulate an immune response to HEL. To assess the contribution of activating and inhibitory FcγR to AMIS, C57BL/6 versus FcRγ-chain(-/-) or FcγRIIB(-/-) mice were used as recipients of HOD-RBC alone or together with anti-HEL Abs (i.e., AMIS) and the resulting immune response to HEL evaluated. We show that anti-HEL polyclonal Abs induce the same degree of AMIS effect in mice lacking these IgG binding receptors as compared with wild-type mice. In agreement with this, F(ab')2 fragments of the AMIS Ab also significantly reduced the Ab response to the HOD cells. In conclusion, successful inhibition of in vivo Ab responses to HOD-RBC by polyclonal IgG can occur independently of activating or inhibitory FcγR involvement. These results may have implications for the understanding of RhD prophylaxis.
Collapse
Affiliation(s)
- Lidice Bernardo
- Canadian Blood Services, Ottawa, Ontario K1G 4J5, Canada; Department of Laboratory Medicine, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada; Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
| | - Honghui Yu
- Canadian Blood Services, Ottawa, Ontario K1G 4J5, Canada; Department of Laboratory Medicine, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada; Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada; Department of Anesthesiology, Tongji Hospital, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Alaa Amash
- Department of Laboratory Medicine, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada; Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
| | - James C Zimring
- Bloodworks Northwest Research Institute, South Lake Union, Seattle, WA 98102
| | - Alan H Lazarus
- Canadian Blood Services, Ottawa, Ontario K1G 4J5, Canada; Department of Laboratory Medicine, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada; Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada; Department of Medicine, University of Toronto, Toronto, Ontario M5S 1A1, Canada; and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A1, Canada
| |
Collapse
|
39
|
Abou Elazab MF, Horiuchi H, Furusawa S. Induction of non-specific suppression in chicks by specific combination of maternal antibody and related antigen. J Vet Med Sci 2015; 77:1363-9. [PMID: 26050841 PMCID: PMC4667651 DOI: 10.1292/jvms.14-0525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Specific immune suppression in newly hatched chicks induced by specific
maternal antibodies has been reported. Laying hens were immunized with
dinitrophenyl-keyhole limpet hemocyanin (DNP-KLH). Purified maternal anti-DNP and
non-specific immunoglobulin (Ig) Y antibodies were transferred by yolk sac inoculation to
newly hatched chicks, and then, they were immunized with an optimum immunogenic dose of
DNP-KLH at 1 and 4 weeks of age. Concentrations of anti-DNP antibodies in serum samples of
these chicks were measured by using Enzyme-linked immunosorbent assay (ELISA). Proportions
of T-cell subsets in peripheral blood of these chicks were also measured by flow
cytometric analysis at 5 weeks of age (one week after the second immunization).
Suppression of anti-DNP antibody response and down-regulation of
CD3+CD4+ cells were observed in the chicks received high dose of
maternal anti-DNP antibodies and immunized with DNP-KLH. On the other hand, normal
anti-DNP antibody response and normal proportion of CD3+CD4+ cells
were observed in the chicks received high dose of non-specific IgY antibodies and
immunized with DNP-KLH. Furthermore, when chicks received high dose of maternal anti-DNP
antibodies and immunized with DNP-KLH at 1 and 4 weeks of age and then with rabbit serum
albumin (RSA) at 5 and 8 weeks of age, their primary anti-RSA response was also
significantly suppressed. We indicate here that specific maternal antibodies can affect
both B and T cell responses and induce non-specific suppression against different
antigens. However, this non-specific suppression does not continue for a long time.
Collapse
Affiliation(s)
- Mohamed Fahmy Abou Elazab
- Laboratory of Immunobiology, Department of Molecular and Applied Bioscience, Graduate School of Biosphere Science, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8528, Japan
| | | | | |
Collapse
|
40
|
Immunization with Immune Complexes Modulates the Fine Specificity of Antibody Responses to a Flavivirus Antigen. J Virol 2015; 89:7970-8. [PMID: 26018152 DOI: 10.1128/jvi.00938-15] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 05/11/2015] [Indexed: 12/30/2022] Open
Abstract
UNLABELLED The antibody response to proteins may be modulated by the presence of preexisting antigen-specific antibodies and the formation of immune complexes (ICs). Effects such as a general increase or decrease of the response as well as epitope-specific phenomena have been described. In this study, we investigated influences of IC immunization on the fine specificity of antibody responses in a structurally well-defined system, using the envelope (E) protein of tick-borne encephalitis (TBE) virus as an immunogen. TBE virus occurs in Europe and Asia and-together with the yellow fever, dengue, West Nile, and Japanese encephalitis viruses-represents one of the major human-pathogenic flaviviruses. Mice were immunized with a dimeric soluble form of E (sE) alone or in complex with monoclonal antibodies specific for each of the three domains of E, and the antibody response induced by these ICs was compared to that seen after immunization with sE alone. Immunoassays using recombinant domains and domain combinations of TBE virus sE as well as the distantly related West Nile virus sE allowed the dissection and quantification of antibody subsets present in postimmunization sera, thus generating fine-specificity patterns of the polyclonal responses. There were substantially different responses with two of the ICs, and the differences could be mechanistically related to (i) epitope shielding and (ii) antibody-mediated structural changes leading to dissociation of the sE dimer. The phenomena described may also be relevant for polyclonal responses upon secondary infections and/or booster immunizations and may affect antibody responses in an individual-specific way. IMPORTANCE Infections with flaviviruses such as yellow fever, dengue, Japanese encephalitis, West Nile, and tick-borne encephalitis (TBE) viruses pose substantial public health problems in different parts of the world. Antibodies to viral envelope protein E induced by natural infection or vaccination were shown to confer protection from disease. Such antibodies can target different epitopes in E protein, and the fine specificities of polyclonal responses can differ between individuals. We conducted a mouse immunization study with TBE E protein alone or complexed to monoclonal antibodies specific for each of the three protein domains. We demonstrated that phenomena such as epitope shielding and antibody-induced structural changes can profoundly influence the fine specificity of antibody responses to the same immunogen. The study thus provided important new information on the potential immunomodulatory role of preexisting antibodies in a flavivirus system that can be relevant for understanding individual-specific factors influencing antibody responses in sequential flavivirus infections and/or immunizations.
Collapse
|
41
|
Gerasimcik N, Dahlberg CIM, Baptista MAP, Massaad MJ, Geha RS, Westerberg LS, Severinson E. The Rho GTPase Cdc42 Is Essential for the Activation and Function of Mature B Cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:4750-8. [PMID: 25870239 DOI: 10.4049/jimmunol.1401634] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 03/16/2015] [Indexed: 12/15/2022]
Abstract
The Rho GTPase Cdc42 coordinates regulation of the actin and the microtubule cytoskeleton by binding and activating the Wiskott-Aldrich syndrome protein. We sought to define the role of intrinsic expression of Cdc42 by mature B cells in their activation and function. Mice with inducible deletion of Cdc42 in mature B cells formed smaller germinal centers and had a reduced Ab response, mostly of low affinity to T cell-dependent Ag, compared with wild-type (WT) controls. Spreading formation of long protrusions that contain F-actin, microtubules, and Cdc42-interacting protein 4, and assumption of a dendritic cell morphology in response to anti-CD40 plus IL-4 were impaired in Cdc42-deficient B cells compared with WT B cells. Cdc42-deficient B cells had an intact migratory response to chemokine in vitro, but their homing to the B cell follicles in the spleen in vivo was significantly impaired. Cdc42-deficient B cells induced a skewed cytokine response in CD4(+) T cells, compared with WT B cells. Our results demonstrate a critical role for Cdc42 in the motility of mature B cells, their cognate interaction with T cells, and their differentiation into Ab-producing cells.
Collapse
Affiliation(s)
- Natalija Gerasimcik
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Carin I M Dahlberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Marisa A P Baptista
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Michel J Massaad
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115; and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115; and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Lisa S Westerberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Eva Severinson
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden;
| |
Collapse
|
42
|
Xu H, van Mechelen L, Henningsson F, Heyman B. Antigen Conjugated to Anti-CD23 Antibodies is Rapidly Transported to Splenic Follicles by Recirculating B Cells. Scand J Immunol 2014; 81:39-45. [DOI: 10.1111/sji.12248] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 10/13/2014] [Indexed: 11/26/2022]
Affiliation(s)
- H. Xu
- Department of Medical Biochemistry and Microbiology; Uppsala University; Uppsala Sweden
| | - L. van Mechelen
- Department of Medical Biochemistry and Microbiology; Uppsala University; Uppsala Sweden
| | - F. Henningsson
- Department of Medical Biochemistry and Microbiology; Uppsala University; Uppsala Sweden
| | - B. Heyman
- Department of Medical Biochemistry and Microbiology; Uppsala University; Uppsala Sweden
| |
Collapse
|
43
|
Dahlberg CIM, He M, Visnes T, Torres ML, Cortizas EM, Verdun RE, Westerberg LS, Severinson E, Ström L. A novel mouse model for the hyper-IgM syndrome: a spontaneous activation-induced cytidine deaminase mutation leading to complete loss of Ig class switching and reduced somatic hypermutation. THE JOURNAL OF IMMUNOLOGY 2014; 193:4732-8. [PMID: 25252954 DOI: 10.4049/jimmunol.1401242] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We describe a spontaneously derived mouse line that completely failed to induce Ig class switching in vitro and in vivo. The mice inherited abolished IgG serum titers in a recessive manner caused by a spontaneous G → A transition mutation in codon 112 of the aicda gene, leading to an arginine to histidine replacement (AID(R112H)). Ig class switching was completely reconstituted by expressing wild-type AID. Mice homozygous for AID(R112H) had peripheral B cell hyperplasia and large germinal centers in the absence of Ag challenge. Immunization with SRBCs elicited an Ag-specific IgG1 response in wild-type mice, whereas AID(R112H) mice failed to produce IgG1 and had reduced somatic hypermutation. The phenotype recapitulates the human hyper-IgM (HIGM) syndrome that is caused by point mutations in the orthologous gene in humans, and the AID(R112H) mutation is frequently found in HIGM patients. The AID(R112H) mouse model for HIGM provides a powerful and more precise tool than conventional knockout strategies.
Collapse
Affiliation(s)
- Carin I M Dahlberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Minghui He
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Torkild Visnes
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; and
| | - Magda Liz Torres
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Elena M Cortizas
- Division of Gerontology and Geriatric Medicine, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Ramiro E Verdun
- Division of Gerontology and Geriatric Medicine, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136
| | - Lisa S Westerberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden;
| | - Eva Severinson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden;
| | - Lena Ström
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; and
| |
Collapse
|
44
|
Niewiesk S. Maternal antibodies: clinical significance, mechanism of interference with immune responses, and possible vaccination strategies. Front Immunol 2014; 5:446. [PMID: 25278941 PMCID: PMC4165321 DOI: 10.3389/fimmu.2014.00446] [Citation(s) in RCA: 343] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 09/01/2014] [Indexed: 01/28/2023] Open
Abstract
Neonates have an immature immune system, which cannot adequately protect against infectious diseases. Early in life, immune protection is accomplished by maternal antibodies transferred from mother to offspring. However, decaying maternal antibodies inhibit vaccination as is exemplified by the inhibition of seroconversion after measles vaccination. This phenomenon has been described in both human and veterinary medicine and is independent of the type of vaccine being used. This review will discuss the use of animal models for vaccine research. I will review clinical solutions for inhibition of vaccination by maternal antibodies, and the testing and development of potentially effective vaccines. These are based on new mechanistic insight about the inhibitory mechanism of maternal antibodies. Maternal antibodies inhibit the generation of antibodies whereas the T cell response is usually unaffected. B cell inhibition is mediated through a cross-link between B cell receptor (BCR) with the Fcγ-receptor IIB by a vaccine-antibody complex. In animal experiments, this inhibition can be partially overcome by injection of a vaccine-specific monoclonal IgM antibody. IgM stimulates the B cell directly through cross-linking the BCR via complement protein C3d and antigen to the complement receptor 2 (CR2) signaling complex. In addition, it was shown that interferon alpha binds to the CD21 chain of CR2 as well as the interferon receptor and that this dual receptor usage drives B cell responses in the presence of maternal antibodies. In lieu of immunizing the infant, the concept of maternal immunization as a strategy to protect neonates has been proposed. This approach would still not solve the question of how to immunize in the presence of maternal antibodies but would defer the time of infection to an age where infection might not have such a detrimental outcome as in neonates. I will review successful examples and potential challenges of implementing this concept.
Collapse
Affiliation(s)
- Stefan Niewiesk
- Department of Veterinary Biosciences, The Ohio State University , Columbus, OH , USA
| |
Collapse
|
45
|
Yu H, Stowell SR, Bernardo L, Hendrickson JE, Zimring JC, Amash A, Uchikawa M, Lazarus AH. Antibody-mediated immune suppression of erythrocyte alloimmunization can occur independently from red cell clearance or epitope masking in a murine model. THE JOURNAL OF IMMUNOLOGY 2014; 193:2902-10. [PMID: 25122924 DOI: 10.4049/jimmunol.1302287] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Anti-D can prevent immunization to the RhD Ag on RBCs, a phenomenon commonly termed Ab-mediated immune suppression (AMIS). The most accepted theory to explain this effect has been the rapid clearance of RBCs. In mouse models using SRBC, these xenogeneic cells are always rapidly cleared even without Ab, and involvement of epitope masking of the SRBC Ags by the AMIS-inducing Ab (anti-SRBC) has been suggested. To address these hypotheses, we immunized mice with murine transgenic RBCs expressing the HOD Ag (hen egg lysozyme [HEL], in sequence with ovalbumin, and the human Duffy transmembrane protein) in the presence of polyclonal Abs or mAbs to the HOD molecule. The isotype, specificity, and ability to induce AMIS of these Abs were compared with accelerated clearance as well as steric hindrance of the HOD Ag. Mice made IgM and IgG reactive with the HEL portion of the molecule only. All six of the mAbs could inhibit the response. The HEL-specific Abs (4B7, IgG1; GD7, IgG2b; 2F4, IgG1) did not accelerate clearance of the HOD-RBCs and displayed partial epitope masking. The Duffy-specific Abs (MIMA 29, IgG2a; CBC-512, IgG1; K6, IgG1) all caused rapid clearance of HOD RBCs without steric hindrance. To our knowledge, this is the first demonstration of AMIS to erythrocytes in an all-murine model and shows that AMIS can occur in the absence of RBC clearance or epitope masking. The AMIS effect was also independent of IgG isotype and epitope specificity of the AMIS-inducing Ab.
Collapse
Affiliation(s)
- Honghui Yu
- The Canadian Blood Services, Ottawa, Ontario K1G 4J5, Canada; Department of Anesthesiology, Tongji Hospital, Huazhong University of Science and Technology, 430030 Wuhan, China; Department of Laboratory Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
| | - Sean R Stowell
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | - Lidice Bernardo
- The Canadian Blood Services, Ottawa, Ontario K1G 4J5, Canada; Department of Laboratory Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
| | | | - James C Zimring
- Puget Sound Blood Center Research Institute, Seattle, WA 98102
| | - Alaa Amash
- Department of Laboratory Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
| | - Makoto Uchikawa
- Kanto-Koshinetsu Block Blood Center, Japanese Red Cross, Koto-ku, Tokyo, Japan 135-8639
| | - Alan H Lazarus
- The Canadian Blood Services, Ottawa, Ontario K1G 4J5, Canada; Department of Laboratory Medicine, Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada; Department of Medicine, University of Toronto, Toronto, Ontario M5S 1A1, Canada; and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A1, Canada
| |
Collapse
|
46
|
Sörman A, Zhang L, Ding Z, Heyman B. How antibodies use complement to regulate antibody responses. Mol Immunol 2014; 61:79-88. [PMID: 25001046 DOI: 10.1016/j.molimm.2014.06.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 06/03/2014] [Accepted: 06/07/2014] [Indexed: 02/06/2023]
Abstract
Antibodies, forming immune complexes with their specific antigen, can cause complete suppression or several 100-fold enhancement of the antibody response. Immune complexes containing IgG and IgM may activate complement and in such situations also complement components will be part of the immune complex. Here, we review experimental data on how antibodies via the complement system upregulate specific antibody responses. Current data suggest that murine IgG1, IgG2a, and IgG2b upregulate antibody responses primarily via Fc-receptors and not via complement. In contrast, IgM and IgG3 act via complement and require the presence of complement receptors 1 and 2 (CR1/2) expressed on both B cells and follicular dendritic cells. Complement plays a crucial role for antibody responses not only to antigen complexed to antibodies, but also to antigen administered alone. Lack of C1q, but not of Factor B or MBL, severely impairs antibody responses suggesting involvement of the classical pathway. In spite of this, normal antibody responses are found in mice lacking several activators of the classical pathway (complement activating natural IgM, serum amyloid P component (SAP), specific intracellular adhesion molecule-grabbing non-integrin R1 (SIGN-R1) or C-reactive protein. Possible explanations to these observations will be discussed.
Collapse
Affiliation(s)
- Anna Sörman
- Department of Medical Biochemistry and Microbiology, Uppsala University, Box 582, BMC, SE-751 23 Uppsala, Sweden
| | - Lu Zhang
- Department of Medical Biochemistry and Microbiology, Uppsala University, Box 582, BMC, SE-751 23 Uppsala, Sweden
| | - Zhoujie Ding
- Department of Medical Biochemistry and Microbiology, Uppsala University, Box 582, BMC, SE-751 23 Uppsala, Sweden
| | - Birgitta Heyman
- Department of Medical Biochemistry and Microbiology, Uppsala University, Box 582, BMC, SE-751 23 Uppsala, Sweden.
| |
Collapse
|
47
|
Abstract
Antibodies in complex with specific antigen can dramatically change the antibody response to this antigen. Depending on antibody class and type of antigen, >99 % suppression or >100-fold enhancement of the response can take place. IgM and IgG3 are efficient enhancers and operate via the complement system. In contrast, IgG1, IgG2a, and IgG2b enhance antibody and CD4(+) T cell responses to protein antigens via activating Fcγ-receptors. IgE also enhances antibody and CD4(+) T cell responses to small proteins but uses the low-affinity receptor for IgE, CD23. Most likely, IgM and IgG3 work by increasing the effective concentration of antigen on follicular dendritic cells in splenic follicles. IgG1, IgG2a, IgG2b, and IgE probably enhance antibody responses by increasing antigen presentation by dendritic cells to T helper cells. IgG antibodies of all subclasses have a dual effect, and suppress antibody responses to particulate antigens such as erythrocytes. This capacity is used in the clinic to prevent immunization of Rhesus-negative women to Rhesus-positive fetal erythrocytes acquired via transplacental hemorrage. IgG-mediated suppression in mouse models can take place in the absence of Fcγ-receptors and complement and to date no knock-out mouse strain has been found where suppression is abrogated.
Collapse
Affiliation(s)
- Birgitta Heyman
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden,
| |
Collapse
|
48
|
Abstract
Antibodies are major molecular effectors of adaptive immune responses. Most, if not all, biological activities of antibodies, however, depend on the functional properties of cells that express receptors for the Fc portion of antibodies (FcR). Most FcR are activating receptors; some are inhibitory. When engaged by antibodies and antigen, the various FcR expressed by a given cell trigger a mixture of positive and negative signals whose integration determines cellular responses. Responses of cell populations can be either protective or pathogenic. As a consequence, FcR are potential target/tools in a variety of diseases including infection, allergy, autoimmune diseases, and cancer.
Collapse
|
49
|
Abstract
Most biological activities of antibodies depend on their ability to engage Receptors for the Fc portion of immunoglobulins (FcRs) on a variety of cell types. As FcRs can trigger positive and negative signals, as these signals control several biological activities in individual cells, as FcRs are expressed by many cells of hematopoietic origin, mostly of the myeloid lineage, as these cells express various combinations of FcRs, and as FcR-expressing cells have different functional repertoires, antibodies can exert a wide spectrum of biological activities. Like B and T Cell Receptors (BCRs and TCRs), FcRs are bona fide immunoreceptors. Unlike BCRs and TCRs, however, FcRs are immunoreceptors with an adaptive specificity for antigen, with an adaptive affinity for antibodies, with an adaptive structure and with an adaptive signaling. They induce adaptive biological responses that depend on their tissue distribution and on FcR-expressing cells that are selected locally by antibodies. They critically determine health and disease. They are thus exquisitely adaptive therapeutic tools.
Collapse
Affiliation(s)
| | - Falk Nimmerjahn
- Department of Biology, Institute of Genetics, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
50
|
Kaufman RM, Schlumpf KS, Wright DJ, Triulzi DJ. Does Rh immune globulin suppress HLA sensitization in pregnancy? Transfusion 2013; 53:2069-77. [PMID: 23252646 PMCID: PMC3609922 DOI: 10.1111/trf.12049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 10/17/2012] [Accepted: 10/30/2012] [Indexed: 12/29/2022]
Abstract
BACKGROUND How Rh immune globulin (RhIG) prevents sensitization to D antigen is unclear. If RhIG Fc delivers a nonspecific immunosuppressive signal, then RhIG may inhibit sensitization to antigens other than D. HLA antibody prevalence was compared in previously pregnant D- versus D+ women to investigate whether RhIG suppresses HLA sensitization. STUDY DESIGN AND METHODS In the Leukocyte Antibody Prevalence Study (LAPS), 7920 volunteer blood donors were screened for anti-HLA and surveyed about prior pregnancies and transfusions. A secondary analysis of the LAPS database was performed. RESULTS D- women not more than 40 years old (presumed to have received antenatal with or without postpartum RhIG in all pregnancies) had a significantly lower HLA sensitization rate than D+ women (relative risk, 0.58; 95% confidence interval [CI], 0.40-0.83). When stratified by deliveries (one, two, three, or four or more), D- women not older than 40 were HLA sensitized less often than D+ women in every case. In contrast, a clear relationship between D type and HLA sensitization was not seen in older previously pregnant women whose childbearing years are presumed to have preceded the use of routine RhIG prophylaxis. In a multivariable logistic regression model, D- women not more than 40 years old remained significantly less likely to be HLA sensitized compared with D+ women after adjusting for parity, time from last pregnancy, lost pregnancies, and transfusions (odds ratio [OR], 0.55; 95% CI, 0.34-0.88). CONCLUSION Consistent with a nonspecific immunosuppressive effect of RhIG, younger previously pregnant D- women were less likely than previously pregnant D+ women to be HLA sensitized.
Collapse
Affiliation(s)
- Richard M Kaufman
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts; Westat, Rockville, Maryland; Institute for Transfusion Medicine, Pittsburgh, Pennsylvania
| | | | | | | |
Collapse
|