1
|
Termote M, Marques RC, Hyllner E, Guryleva MV, Henskens M, Brutscher A, Baken IJL, Dopico XC, Gasull AD, Murrell B, Stamatatos L, Westerberg LS, Dosenovic P. Antigen affinity and site of immunization dictate B cell recall responses. Cell Rep 2025; 44:115221. [PMID: 39817910 DOI: 10.1016/j.celrep.2024.115221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 11/29/2024] [Accepted: 12/26/2024] [Indexed: 01/18/2025] Open
Abstract
Protective antibodies against HIV-1 require unusually high levels of somatic mutations introduced in germinal centers (GCs). To achieve this, a sequential vaccination approach was proposed. Using HIV-1 antibody knockin mice with fate-mapping genes, we examined if antigen affinity affects the outcome of B cell recall responses. Compared to a high-affinity boost, a low-affinity boost resulted in decreased numbers of memory-derived B cells in secondary GCs but with higher average levels of somatic mutations, indicating an affinity threshold for memory B cells to enter GCs. Furthermore, upon boosting local lymph nodes (LNs), the composition of primary GCs was modified in an antigen-affinity-dependent manner to constitute less somatically mutated B cells. Our results demonstrate that antigen affinity and location of the boost affect the outcome of the B cell recall response. These results can help guide the design of vaccine immunogens aiming to selectively engage specific B cell clones for further diversification.
Collapse
Affiliation(s)
- Manon Termote
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Rafael C Marques
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Erik Hyllner
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Mariia V Guryleva
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Mirthe Henskens
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Andreas Brutscher
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Isabel J L Baken
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Xaquin Castro Dopico
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Adria Dalmau Gasull
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Ben Murrell
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Leonidas Stamatatos
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Lisa S Westerberg
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden
| | - Pia Dosenovic
- Department of Microbiology, Tumor and Cell Biology, Division of Virology and Immunology, Karolinska Institutet, 171 65 Solna, Sweden.
| |
Collapse
|
2
|
Mei HE, Schulz AR, Hirseland H, Diekmann LM, Habermann E, Ten Hagen A, Albach F, Burmester GR, Grützkau A, Biesen R. Prophylactic antibodies inhibit spike-specific T and B cell responses after COVID-19 vaccination. J Med Virol 2024; 96:e29778. [PMID: 38965882 DOI: 10.1002/jmv.29778] [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: 12/28/2023] [Revised: 04/29/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024]
Abstract
Active and passive immunization is used in high-risk patients to prevent severe courses of COVID-19, but the impact of prophylactic neutralizing antibodies on the immune reaction to the mRNA vaccines has remained enigmatic. Here we show that CD4 T and B cell responses to Spikevax booster immunization are suppressed by the therapeutic antibodies Casirivimab and Imdevimab. B cell and T cell responses were significantly induced in controls but not in antibody-treated patients. The data indicates that humoral immunity, i. e. high levels of antibodies, negatively impacts reactive immunity, resulting in blunted cellular responses upon boosting. This argues for temporal separation of vaccination efforts; with active vaccination preferably applied before prophylactic therapeutic antibody treatment.
Collapse
MESH Headings
- Humans
- COVID-19/prevention & control
- COVID-19/immunology
- B-Lymphocytes/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- COVID-19 Vaccines/immunology
- SARS-CoV-2/immunology
- Middle Aged
- Male
- Female
- Vaccination
- Adult
- Aged
- CD4-Positive T-Lymphocytes/immunology
- T-Lymphocytes/immunology
- Immunization, Secondary
- Immunity, Humoral
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/therapeutic use
Collapse
Affiliation(s)
- Henrik E Mei
- Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Axel Ronald Schulz
- Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Heike Hirseland
- Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Lisa-Marie Diekmann
- Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Elisa Habermann
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alexander Ten Hagen
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Fredrik Albach
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gerd-Rüdiger Burmester
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Grützkau
- Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Robert Biesen
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
3
|
Beneduce C, Nguyen S, Washburn N, Schaeck J, Meccariello R, Holte K, Ortiz D, Manning AM, Bosques CJ, Kurtagic E. Inhibitory Fc-Gamma IIb Receptor Signaling Induced by Multivalent IgG-Fc Is Dependent on Sialylation. Cells 2023; 12:2130. [PMID: 37681862 PMCID: PMC10486564 DOI: 10.3390/cells12172130] [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: 07/11/2023] [Revised: 08/05/2023] [Accepted: 08/18/2023] [Indexed: 09/09/2023] Open
Abstract
Immunoglobulin (IgG) Fc glycosylation has been shown to be important for the biological activity of antibodies. Fc sialylation is important for the anti-inflammatory activity of IgGs. However, evaluating the structure-activity relationship (SAR) of antibody Fc glycosylation has been hindered using simplified in vitro models in which antibodies are often displayed in monomeric forms. Presenting antibodies in monomeric forms may not accurately replicate the natural environment of the antibodies when binding their antigen in vivo. To address these limitations, we used different Fc-containing molecules, displaying their Fc domains in monovalent and multivalent fashion. Given the inhibitory role of Fc gamma receptor IIb (FcγRIIb) in autoimmune and inflammatory diseases, we focused on evaluating the impact of Fc sialylation on the activation of FcγRIIb. We report for the first time that in human cellular systems, sialic acid mediates the induction of FcγRIIb phosphorylation by IgG-Fc when the IgG-Fc is displayed in a multivalent fashion. This effect was observed with different types of therapeutic agents such as sialylated anti-TNFα antibodies, sialylated IVIg and sialylated recombinant multivalent Fc products. These studies represent the first report of the specific effects of Fc sialylation on FcγRIIb signaling on human immune cells and may help in the characterization of the anti-inflammatory activity of Fc-containing therapeutic candidates.
Collapse
Affiliation(s)
- Christopher Beneduce
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
- Janssen Research & Development, Cambridge, MA 02142, USA
| | | | - Nathaniel Washburn
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
- Janssen Research & Development, Cambridge, MA 02142, USA
| | - John Schaeck
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
| | - Robin Meccariello
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
- Janssen Research & Development, Cambridge, MA 02142, USA
| | | | - Daniel Ortiz
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
| | | | | | - Elma Kurtagic
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
- Janssen Research & Development, Cambridge, MA 02142, USA
| |
Collapse
|
4
|
Chevillard C, Amen A, Besson S, Hannani D, Bally I, Dettling V, Gout E, Moreau CJ, Buisson M, Gallet S, Fenel D, Vassal-Stermann E, Schoehn G, Poignard P, Dagher MC, Fender P. Elicitation of potent SARS-CoV-2 neutralizing antibody responses through immunization with a versatile adenovirus-inspired multimerization platform. Mol Ther 2022; 30:1913-1925. [PMID: 35151843 PMCID: PMC8828441 DOI: 10.1016/j.ymthe.2022.02.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/19/2022] [Accepted: 02/07/2022] [Indexed: 11/24/2022] Open
Abstract
Virus-like particles (VLPs) are highly suited platforms for protein-based vaccines. In the present work, we adapted a previously designed non-infectious adenovirus-inspired 60-mer dodecahedric VLP (ADDomer) to display a multimeric array of large antigens through a SpyTag/SpyCatcher system. To validate the platform as a potential COVID-19 vaccine approach, we decorated the newly designed VLP with the glycosylated receptor binding domain (RBD) of SARS-CoV-2. Cryoelectron microscopy structure revealed that up to 60 copies of this antigenic domain could be bound on a single ADDomer particle, with the symmetrical arrangements of a dodecahedron. Mouse immunization with the RBD decorated VLPs already showed a significant specific humoral response following prime vaccination, greatly reinforced by a single boost. Neutralization assays with SARS-CoV-2 spike pseudo-typed virus demonstrated the elicitation of strong neutralization titers, superior to those of COVID-19 convalescent patients. Notably, the presence of pre-existing immunity against the adenoviral-derived particles did not hamper the immune response against the antigen displayed on its surface. This plug and play vaccine platform represents a promising new highly versatile tool to combat emergent pathogens.
Collapse
Affiliation(s)
- Christopher Chevillard
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Axelle Amen
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France; CHU Grenoble Alpes, 38000 Grenoble, France
| | - Solène Besson
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Dalil Hannani
- University Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC, 38000 Grenoble, France
| | - Isabelle Bally
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Valentin Dettling
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Evelyne Gout
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Christophe J Moreau
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Marlyse Buisson
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Salomé Gallet
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Daphna Fenel
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Emilie Vassal-Stermann
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Guy Schoehn
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Pascal Poignard
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France; CHU Grenoble Alpes, 38000 Grenoble, France; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Marie-Claire Dagher
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France
| | - Pascal Fender
- CNRS, Univ. Grenoble Alpes, CEA, UMR5075, Institut de Biologie Structurale, 38042 Grenoble, France.
| |
Collapse
|
5
|
Baumrucker CR, Macrina AL, Bruckmaier RM. Colostrogenesis: Role and Mechanism of the Bovine Fc Receptor of the Neonate (FcRn). J Mammary Gland Biol Neoplasia 2021; 26:419-453. [PMID: 35080749 DOI: 10.1007/s10911-021-09506-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 12/10/2021] [Indexed: 11/28/2022] Open
Abstract
Colostrogenesis is a separate and unique phase of mammary epithelial cell activity occurring in the weeks before parturition and rather abruptly ending after birth in the bovine. It has been the focus of research to define what controls this process and how it produces high concentrations of specific biologically active components important for the neonate. In this review we consider colostrum composition and focus upon components that appear in first milked colostrum in concentrations exceeding that in blood serum. The Fc Receptor of the Neonate (FcRn) is recognized as the major immunoglobulin G (IgG) and albumin binding protein that accounts for the proteins' long half-lives. We integrate the action of the pinocytotic (fluid phase) uptake of extracellular components and merge them with FcRn in sorting endosomes. We define and explore the means of binding, sorting, and the transcytotic delivery of IgG1 while recycling IgG2 and albumin. We consider the means of releasing the ligands from the receptor within the endosome and describe a new secretion mechanism of cargo release into colostrum without the appearance of FcRn itself in colostrum. We integrate the insulin-like growth factor family, some of which are highly concentrated bioactive components of colostrum, with the mechanisms related to FcRn endosome action. In addition to secretion, we highlight the recent findings of a role of the FcRn in phagocytosis and antigen presentation and relate its significant and abrupt change in cellular location after parturition to a role in the prevention and resistance to mastitis infections.
Collapse
Affiliation(s)
- Craig R Baumrucker
- Department of Animal Science, Penn State University, University Park, PA, 16802, USA.
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland.
| | - Ann L Macrina
- Department of Animal Science, Penn State University, University Park, PA, 16802, USA
| | - Rupert M Bruckmaier
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland
| |
Collapse
|
6
|
Clemens EA, Alexander-Miller MA. Understanding Antibody Responses in Early Life: Baby Steps towards Developing an Effective Influenza Vaccine. Viruses 2021; 13:v13071392. [PMID: 34372597 PMCID: PMC8310046 DOI: 10.3390/v13071392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/13/2021] [Indexed: 01/01/2023] Open
Abstract
The immune system of young infants is both quantitatively and qualitatively distinct from that of adults, with diminished responsiveness leaving these individuals vulnerable to infection. Because of this, young infants suffer increased morbidity and mortality from respiratory pathogens such as influenza viruses. The impaired generation of robust and persistent antibody responses in these individuals makes overcoming this increased vulnerability through vaccination challenging. Because of this, an effective vaccine against influenza viruses in infants under 6 months is not available. Furthermore, vaccination against influenza viruses is challenging even in adults due to the high antigenic variability across viral strains, allowing immune evasion even after induction of robust immune responses. This has led to substantial interest in understanding how specific antibody responses are formed to variable and conserved components of influenza viruses, as immune responses tend to strongly favor recognition of variable epitopes. Elicitation of broadly protective antibody in young infants, therefore, requires that both the unique characteristics of young infant immunity as well as the antibody immunodominance present among epitopes be effectively addressed. Here, we review our current understanding of the antibody response in newborns and young infants and discuss recent developments in vaccination strategies that can modulate both magnitude and epitope specificity of IAV-specific antibody.
Collapse
|
7
|
Hu C, Peng S, Zhao L, Li M, Liu M, Xu Y, Chen G. Yishen-tongbi decoction inhibits excessive activation of B cells by activating the FcγRIIb/Lyn/SHP-1 pathway and attenuates the inflammatory response in CIA rats. Biomed Pharmacother 2021; 134:111166. [PMID: 33373915 DOI: 10.1016/j.biopha.2020.111166] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/17/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease. Strong evidence supports that excessive activation of B cells plays a critical role in the pathogenesis of RA. Fc gamma receptor b (FcγRIIb) is the B cell inhibitory receptor and inhibits BCR (B cell receptor) signalling in part by selectively dephosphorylating CD19 which is considered a co-receptor for BCR and is essential for B cell activation. Our previous study demonstrated that a FcγRIIb I232T polymorphism presented a strong genetic link to RA and may lead to the excessive activation of B cells. Therefore, novel therapeutic strategies and drugs that can effectively inhibit the excessive activation of B cells by regulating the FcγRIIb are necessary for the treatment of RA. Therefore, we used Burkitt's lymphoma ST486 human B cells (lacking endogenous FcγRIIb) transfected with the 232Thr loss-of-function mutant to construct a FcγRIIb mutant cell line (ST486), and we demonstrated that YSTB treatment not only reduced proliferation and promoted apoptosis in ST486 cells but also did so in a dose-dependent manner. Furthermore, the intracellular Ca2+ flux of ST486 cells was decreased after treatment with YSTB, inhibiting the excessive activation of ST486 cells, and these effects correlated with the CD19/FcγRIIb-Lyn-SHP-1 pathways. Our data showed that YSTB treatment inhibited the expression of phosphorylated CD19 and upregulated the protein expression of FcγRIIb, Lyn, and SHP-1. Additionally, the CIA model was established to explore the anti-inflammatory and inhibitory effects of YSTB on bone destruction, and we found that YSTB decreased the paw oedema and arthritis index (AI) in CIA rats. It is worth mentioning that YSTB clearly decreased the AI earlier than methotrexate (MTX) (day 10 vs 16). Moreover, synovial hyperplasia, inflammatory cell infiltration and cartilage surface erosion in CIA rats were noticeably reduced after treatment with YSTB as evidenced by histopathological examination. Finally, we found that YSTB treatment suppressed bone erosion and joint space score (JNS) in CIA rats as evidenced by radiographic assessment. In summary, these data suggest that YSTB has great therapeutic potential for RA treatment.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/pharmacology
- Apoptosis/drug effects
- Arthritis, Experimental/chemically induced
- Arthritis, Experimental/immunology
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/prevention & control
- B-Lymphocytes/drug effects
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Collagen Type II
- Drugs, Chinese Herbal/pharmacology
- Female
- Humans
- Joints/drug effects
- Joints/immunology
- Joints/metabolism
- Joints/pathology
- Lymphocyte Activation/drug effects
- Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism
- Rats, Wistar
- Receptors, IgG/genetics
- Receptors, IgG/metabolism
- Signal Transduction
- src-Family Kinases/genetics
- src-Family Kinases/metabolism
- Rats
Collapse
Affiliation(s)
- Congqi Hu
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shanqin Peng
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lianyu Zhao
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meilin Li
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Muqiu Liu
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanping Xu
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangxing Chen
- Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Baiyun Hospital of The First Affiliated Hospital of Guangzhou University of Chinese Medicine, China.
| |
Collapse
|
8
|
Challenges for the Newborn Immune Response to Respiratory Virus Infection and Vaccination. Vaccines (Basel) 2020; 8:vaccines8040558. [PMID: 32987691 PMCID: PMC7712002 DOI: 10.3390/vaccines8040558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/19/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
The initial months of life reflect an extremely challenging time for newborns as a naïve immune system is bombarded with a large array of pathogens, commensals, and other foreign entities. In many instances, the immune response of young infants is dampened or altered, resulting in increased susceptibility and disease following infection. This is the result of both qualitative and quantitative changes in the response of multiple cell types across the immune system. Here we provide a review of the challenges associated with the newborn response to respiratory viral pathogens as well as the hurdles and advances for vaccine-mediated protection.
Collapse
|
9
|
Dhenni R, Phan TG. The geography of memory B cell reactivation in vaccine-induced immunity and in autoimmune disease relapses. Immunol Rev 2020; 296:62-86. [PMID: 32472583 DOI: 10.1111/imr.12862] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/05/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022]
Abstract
Memory B cells (Bmem) provide an active second layer of defense against re-infection by pathogens that have bypassed the passive first layer provided by neutralizing antibodies. Here, we review recent progress in our understanding of Bmem heterogeneity in terms of their origin (germinal center-dependent vs center-independent), phenotype (canonical vs atypical vs age-associated B cells), trafficking (recirculating vs tissue-resident), and fate (plasma cell vs germinal center differentiation). The development of transgenic models and intravital imaging technologies has made it possible to track the cellular dynamics of Bmem reactivation by antigen, their interactions with follicular memory T cells, and differentiation into plasma cells in subcapsular proliferative foci in the lymph nodes of immune animals. Such in situ studies have reinforced the importance of geography in shaping the outcome of the secondary antibody response. We also review the evidence for Bmem reactivation and differentiation into short-lived plasma cells in the pathogenesis of disease flares in relapsing-remitting autoimmune diseases. Elucidating the mechanisms that control the Bmem fate decision to differentiate into plasma cells or germinal center B cells will aid future efforts to more precisely engineer fit-for-purpose vaccines as well as to treat antibody-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Rama Dhenni
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Tri Giang Phan
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
10
|
Dhamodharan U, Teena R, Vimal Kumar R, Changam SS, Ramkumar KM, Rajesh K. Circulatory levels of B-cell activating factor of the TNF family in patients with diabetic foot ulcer: Association with disease progression. Wound Repair Regen 2019; 27:442-449. [PMID: 31041853 DOI: 10.1111/wrr.12720] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 03/15/2019] [Accepted: 04/01/2019] [Indexed: 11/29/2022]
Abstract
Enhanced and prolonged expression of tumor necrosis factor alpha (TNF-α), a potent pro-inflammatory cytokine is evidenced during the chronic wound healing process of infected diabetic foot ulcer (IDFU). B-cell activating factor (BAFF) is the member of TNF-α family, which implicit in B-cell dysfunction. This study was aimed to evaluate the role of BAFF in diabetic foot ulcer (DFU) patients and to correlate its association with other family of inflammatory cytokines. Circulating levels of BAFF and other cytokines were measured in IDFU (n = 44) and non-IDFU patients (n = 40) using multiplexed bead-based cytokine immunoassay. A stepwise significant increase was observed in both circulatory BAFF and C-reactive protein (CRP) during the disease progression. The area under the receiver operating characteristic curve (AUCROC ) for BAFF was found to be high (0.89; [95% CI: 0.73-1.0]), when compared to CRP (0.68; [95% CI: 0.61-0.76]). Optimum diagnostic cutoff level for BAFF was found to be ≥2.35 pg/mL with 62.0% sensitivity and 85.7% specificity. Further, BAFF levels showed a significant positive correlation with CRP among IDFU patients. With respect to other family cytokines, BAFF levels were positively correlated with TNF-α, interferon family cytokines such as IFN-α2, IL-28A/IFN-λ2, IFN-γ, and IL-10 family cytokines such as IL-19, IL-22, and IL-26 and negatively correlated with IL-6 receptor family such as gp130/sIL-6Rβ. Hence, our data suggest that devising therapeutic strategies to reduce the levels of BAFF may contribute in amelioration of IDFU.
Collapse
Affiliation(s)
- Umapathy Dhamodharan
- Life Science Division, SRM Research Institute, SRM University, Kattankulathur, 603 203, Tamil Nadu, India
| | - Rajan Teena
- Department of Biotechnology, School of Bio-engineering, SRM University, Kattankulathur, 603 203, Tamil Nadu, India
| | - Ramachandran Vimal Kumar
- Department of Podiatry, Hycare Super Speciality Hospital, MMDA Colony, Arumbakkam, Chennai, 600 106, Tamil Nadu, India
| | - Sheela Sasikumar Changam
- Department of Podiatry, Hycare Super Speciality Hospital, MMDA Colony, Arumbakkam, Chennai, 600 106, Tamil Nadu, India
| | - Kunka Mohanram Ramkumar
- Life Science Division, SRM Research Institute, SRM University, Kattankulathur, 603 203, Tamil Nadu, India.,Department of Biotechnology, School of Bio-engineering, SRM University, Kattankulathur, 603 203, Tamil Nadu, India
| | - Kesavan Rajesh
- Department of Podiatry, Hycare Super Speciality Hospital, MMDA Colony, Arumbakkam, Chennai, 600 106, Tamil Nadu, India
| |
Collapse
|
11
|
|
12
|
Okada N, Sasaki A, Saito J, Mitani Y, Yachie A, Takahashi H, Matsubara S, Tenkumo C, Tanaka H, Hata T, Motomura K, Nagasawa J, Wada Y, Sako M, Yamaguchi K, Matsumoto K, Nakamura H, Sago H, Mizuta K. The Japanese experience and pharmacokinetics of antenatal maternal high-dose immunoglobulin treatment as a prophylaxis for neonatal hemochromatosis in siblings. J Matern Fetal Neonatal Med 2018; 33:142-148. [PMID: 29890876 DOI: 10.1080/14767058.2018.1487940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Background: Neonatal hemochromatosis (NH) is a rare but serious disease causing fulminant hepatic failure. The recurrence rate of NH in a subsequent infant of a mother with an affected infant is 70-90%. Recently, antenatal maternal high-dose intravenous immunoglobulin (IVIG) treatment has been reported to be effective for preventing NH recurrence. However, data on the IgG concentrations during this treatment are limited.Objective: We report a Japanese experience and present a pharmacokinetic simulation model of IgG during IVIG treatment.Methods: Women with histories of pregnancy diagnosed with NH were treated with IVIG weekly from the second trimester until the end of gestation. Serum IgG levels during treatment were collected frequently and pharmacokinetics were simulated by a two-compartment model.Results: Six women were included during eight pregnancies. None experienced severe adverse events. Three out of eight infants showed temporary liver dysfunction, but none required any treatment. A simulation study showed that the estimated trough and peak levels of IgG concentrations during IVIG were 2000-3000 and 4000-5000 mg/dl, respectively.Conclusion: This treatment prevented the recurrence of NH in siblings in Japanese women. We examined the details of serum IgG concentrations and introduced a new pharmacokinetic simulation model of IgG concentrations during IVIG treatment.
Collapse
Affiliation(s)
- Noriki Okada
- Department of Transplant Surgery, Jichi Medical University, Shimotsuke, Japan
| | - Aiko Sasaki
- Center of Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Division of Obstetrics, Tokyo, Japan
| | - Jumpei Saito
- Department of Pharmacy, National Center for Child Health and Development, Tokyo, Japan
| | - Yusuke Mitani
- Perinatal Mother and Infant Care Center, Kanazawa University Hospital, Kanazawa, Japan
| | - Akihiro Yachie
- Department of Pediatrics, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Hironori Takahashi
- Department of Obstetrics and Gynecology, Jichi Medical University, Shimotsuke, Japan
| | - Shigeki Matsubara
- Department of Obstetrics and Gynecology, Jichi Medical University, Shimotsuke, Japan
| | - Chiaki Tenkumo
- Department of Perinatology and Gynecology, Kagawa University Graduate School of Medicine, Miki, Japan
| | - Hirokazu Tanaka
- Department of Perinatology and Gynecology, Kagawa University Graduate School of Medicine, Miki, Japan
| | - Toshiyuki Hata
- Department of Perinatology and Gynecology, Kagawa University Graduate School of Medicine, Miki, Japan
| | - Kenichiro Motomura
- Department of Allergy and Clinical Immunology, National Center for Child Health and Development, Tokyo, Japan
| | - Junko Nagasawa
- Center of Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Division of Obstetrics, Tokyo, Japan
| | - Yuka Wada
- Center of Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Division of Obstetrics, Tokyo, Japan
| | - Mayumi Sako
- Division for Clinical Trials, Department of Clinical Research, Center for Clinical Research and Development, National Center for Child Health and Development, Tokyo, Japan
| | - Koshi Yamaguchi
- Center of Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Division of Obstetrics, Tokyo, Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Center for Child Health and Development, Tokyo, Japan
| | - Hidefumi Nakamura
- Department of Development Strategy, National Center for Child Health and Development, Tokyo, Japan
| | - Haruhiko Sago
- Center of Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Division of Obstetrics, Tokyo, Japan
| | - Koichi Mizuta
- Department of Transplant Surgery, Jichi Medical University, Shimotsuke, Japan
| |
Collapse
|
13
|
Forsthuber TG, Cimbora DM, Ratchford JN, Katz E, Stüve O. B cell-based therapies in CNS autoimmunity: differentiating CD19 and CD20 as therapeutic targets. Ther Adv Neurol Disord 2018; 11:1756286418761697. [PMID: 29593838 PMCID: PMC5865455 DOI: 10.1177/1756286418761697] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/01/2018] [Indexed: 01/05/2023] Open
Abstract
Increasing recognition of the role of B cells in the adaptive immune response makes B cells an important therapeutic target in autoimmunity. Numerous current and developmental immunotherapies target B cells for elimination through recognition of cell-surface proteins expressed specifically on B cells, in particular CD19 and CD20. Similarities and differences in the function and expression of these two molecules predict some shared, and some distinct, pharmacological effects of agents targeting CD19 versus CD20, potentially leading to differences in the clinical safety and efficacy of such agents. Here, we review current knowledge of CD19 and CD20 function and biology, survey current and developmental therapies that target these molecules, and discuss potential differences in elimination of B cells by drugs that target CD19 versus CD20, with particular focus on the central nervous system autoimmune diseases multiple sclerosis and neuromyelitis optica. The principles and mechanisms herein discussed might also be relevant to a variety of other nervous system autoimmune disorders, including NMDA (N-methyl-D-aspartate) receptor encephalitis, transverse myelitis and myasthenia gravis.
Collapse
Affiliation(s)
| | | | | | | | - Olaf Stüve
- Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, TX, USA
| |
Collapse
|
14
|
Stopforth RJ, Oldham RJ, Tutt AL, Duriez P, Chan HTC, Binkowski BF, Zimprich C, Li D, Hargreaves PG, Cong M, Reddy V, Leandro MJ, Cambridge G, Lux A, Nimmerjahn F, Cragg MS. Detection of Experimental and Clinical Immune Complexes by Measuring SHIP-1 Recruitment to the Inhibitory FcγRIIB. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:1937-1950. [PMID: 29351998 PMCID: PMC5837011 DOI: 10.4049/jimmunol.1700832] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 12/20/2017] [Indexed: 12/16/2022]
Abstract
Fc γ receptors (FcγR) are involved in multiple aspects of immune cell regulation, are central to the success of mAb therapeutics, and underpin the pathology of several autoimmune diseases. However, reliable assays capable of accurately measuring FcγR interactions with their physiological ligands, IgG immune complexes (IC), are limited. A method to study and detect IC interactions with FcγRs was therefore developed. This method, designed to model the signaling pathway of the inhibitory FcγRIIB (CD32B), used NanoLuc Binary Interaction Technology to measure recruitment of the Src homology 2 domain-containing inositol phosphatase 1 to the ITIM of this receptor. Such recruitment required prior cross-linking of an ITAM-containing activatory receptor, and evoked luciferase activity in discrete clusters at the cell surface, recapitulating the known biology of CD32B signaling. The assay detected varying forms of experimental IC, including heat-aggregated IgG, rituximab-anti-idiotype complexes, and anti-trinitrophenol-trinitrophenol complexes in a sensitive manner (≤1 μg/ml), and discriminated between complexes of varying size and isotype. Proof-of-concept for the detection of circulating ICs in autoimmune disease was provided, as responses to sera from patients with systemic lupus erythematosus and rheumatoid arthritis were detected in small pilot studies. Finally, the method was translated to a stable cell line system. In conclusion, a rapid and robust method for the detection of IC was developed, which has numerous potential applications including the monitoring of IC in autoimmune diseases and the study of underlying FcγR biology.
Collapse
Affiliation(s)
- Richard J Stopforth
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| | - Robert J Oldham
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| | - Alison L Tutt
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| | - Patrick Duriez
- Southampton Experimental Cancer Medicine/Cancer Research U.K. Centre, Protein Core Facility, Cancer Sciences Unit, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| | - H T Claude Chan
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| | | | | | - Dun Li
- Promega Corp., Fitchburg, WI 53711
| | - Philip G Hargreaves
- Promega UK Ltd., Southampton Science Park, Southampton SO16 7NS, United Kingdom
| | - Mei Cong
- Promega Corp., Fitchburg, WI 53711
| | - Venkat Reddy
- Division of Medicine, Centre for Rheumatology, University College London, London WC1E 6JF, United Kingdom; and
| | - Maria J Leandro
- Division of Medicine, Centre for Rheumatology, University College London, London WC1E 6JF, United Kingdom; and
| | - Geraldine Cambridge
- Division of Medicine, Centre for Rheumatology, University College London, London WC1E 6JF, United Kingdom; and
| | - Anja Lux
- Department of Biology, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Falk Nimmerjahn
- Department of Biology, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Mark S Cragg
- Antibody and Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, United Kingdom;
| |
Collapse
|
15
|
Roghanian A, Stopforth RJ, Dahal LN, Cragg MS. New revelations from an old receptor: Immunoregulatory functions of the inhibitory Fc gamma receptor, FcγRIIB (CD32B). J Leukoc Biol 2018; 103:1077-1088. [PMID: 29406570 DOI: 10.1002/jlb.2mir0917-354r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/03/2017] [Accepted: 12/14/2017] [Indexed: 12/11/2022] Open
Abstract
The Fc gamma receptor IIB (FcγRIIB/CD32B) was generated million years ago during evolution. It is the sole inhibitory receptor for IgG, and has long been associated with the regulation of humoral immunity and innate immune homeostasis. However, new and surprising functions of FcγRIIB are emerging. In particular, FcγRIIB has been shown to perform unexpected activatory roles in both immune-signaling and monoclonal antibody (mAb) immunotherapy. Furthermore, although ITIM signaling is an integral part of FcγRIIB regulatory activity, it is now clear that inhibition/activation of immune responses can occur independently of the ITIM. In light of these new findings, we present an overview of the established and noncanonical functions of FcγRIIB and discuss how this knowledge might be exploited therapeutically.
Collapse
Affiliation(s)
- Ali Roghanian
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Richard J Stopforth
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Lekh N Dahal
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Mark S Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| |
Collapse
|
16
|
Lin Y, Song C, Xu J, Yang Y, Qin H, Zhao C, Lin J, Liu R, Li Z. Irrelevance of anti-nAChR cytoplasmic loop antibody in the degree of myasthenia gravis. Int J Mol Med 2017; 40:931-939. [DOI: 10.3892/ijmm.2017.3074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 07/10/2017] [Indexed: 11/05/2022] Open
|
17
|
Fejfarová V, Jirkovská A, Dubský M, Game F, Vydláková J, Sekerková A, Franeková J, Kučerová M, Stříž I, Petkov V, Bém R, Wosková V, Němcová A, Skibová J. An Alteration of Lymphocytes Subpopulations and Immunoglobulins Levels in Patients with Diabetic Foot Ulcers Infected Particularly by Resistant Pathogens. J Diabetes Res 2016; 2016:2356870. [PMID: 28050566 PMCID: PMC5165150 DOI: 10.1155/2016/2356870] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 09/30/2016] [Accepted: 10/17/2016] [Indexed: 01/16/2023] Open
Abstract
The aim of our study was to analyse immune abnormalities in patients with chronic infected diabetic foot ulcers (DFUs) especially those infected by resistant microorganisms. Methods. 68 patients treated in our foot clinic for infected chronic DFUs with 34 matched diabetic controls were studied. Patients with infected DFUs were subdivided into two subgroups according to the antibiotic sensitivity of causal pathogen: subgroup S infected by sensitive (n = 50) and subgroup R by resistant pathogens (n = 18). Selected immunological markers were compared between the study groups and subgroups. Results. Patients with infected chronic DFUs had, in comparison with diabetic controls, significantly reduced percentages (p < 0.01) and total numbers of lymphocytes (p < 0.001) involving B lymphocytes (p < 0.01), CD4+ (p < 0.01), and CD8+ T cells (p < 0.01) and their naive and memory effector cells. Higher levels of IgG (p < 0.05) including IgG1 (p < 0.001) and IgG3 (p < 0.05) were found in patients with DFUs compared to diabetic controls. Serum levels of immunoglobulin subclasses IgG2 and IgG3 correlated negatively with metabolic control (p < 0.05). A trend towards an increased frequency of IgG2 deficiency was found in patients with DFUs compared to diabetic controls (22% versus 15%; NS). Subgroup R revealed lower levels of immunoglobulins, especially of IgG4 (p < 0.01) in contrast to patients infected by sensitive bacteria. The innate immunity did not differ significantly between the study groups. Conclusion. Our study showed changes mainly in the adaptive immune system represented by low levels of lymphocyte subpopulations and their memory effector cells, and also changes in humoral immunity in patients with DFUs, even those infected by resistant pathogens, in comparison with diabetic controls.
Collapse
Affiliation(s)
- Vladimíra Fejfarová
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Alexandra Jirkovská
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Michal Dubský
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Frances Game
- Diabetes Unit, Derby Hospitals NHS Foundation Trust, Derby, UK
| | - Jana Vydláková
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Alena Sekerková
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jana Franeková
- Department of Clinical Biochemistry, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Monika Kučerová
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ilja Stříž
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Vladimír Petkov
- Department of Clinical Microbiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Robert Bém
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Veronika Wosková
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Andrea Němcová
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jelena Skibová
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
18
|
Control of memory B cell responses by extrinsic and intrinsic mechanisms. Immunol Lett 2016; 178:27-30. [DOI: 10.1016/j.imlet.2016.05.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 05/16/2016] [Indexed: 11/23/2022]
|
19
|
Stapleton NM, Einarsdóttir HK, Stemerding AM, Vidarsson G. The multiple facets of FcRn in immunity. Immunol Rev 2016; 268:253-68. [PMID: 26497526 DOI: 10.1111/imr.12331] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The neonatal Fc receptor, FcRn, is best known for its role in transporting IgG in various tissues, providing newborns with humoral immunity, and for prolonging the half-life of IgG. Recent findings implicate the involvement of FcRn in a far wider range of biological and immunological processes, as FcRn has been found to bind and extend the half-life of albumin; to be involved in IgG transport and antigen sampling at mucosal surfaces; and to be crucial for efficient IgG-mediated phagocytosis. Herein, the function of FcRn will be reviewed, with emphasis on its recently documented significance for IgG polymorphisms affecting the half-life and biodistribution of IgG3, on its role in phagocyte biology, and the subsequent role for the presentation of antigens to lymphocytes.
Collapse
Affiliation(s)
- Nigel M Stapleton
- Sanquin Research and Landsteiner Laboratory, Amsterdam Medical Centre, Amsterdam, The Netherlands
| | - Helga K Einarsdóttir
- Sanquin Research and Landsteiner Laboratory, Amsterdam Medical Centre, Amsterdam, The Netherlands
| | | | - Gestur Vidarsson
- Sanquin Research and Landsteiner Laboratory, Amsterdam Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Pauls SD, Ray A, Hou S, Vaughan AT, Cragg MS, Marshall AJ. FcγRIIB-Independent Mechanisms Controlling Membrane Localization of the Inhibitory Phosphatase SHIP in Human B Cells. THE JOURNAL OF IMMUNOLOGY 2016; 197:1587-96. [PMID: 27456487 DOI: 10.4049/jimmunol.1600105] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 06/21/2016] [Indexed: 01/08/2023]
Abstract
SHIP is an important regulator of immune cell signaling that functions to dephosphorylate the phosphoinositide phosphatidylinositol 3,4,5-trisphosphate at the plasma membrane and mediate protein-protein interactions. One established paradigm for SHIP activation involves its recruitment to the phospho-ITIM motif of the inhibitory receptor FcγRIIB. Although SHIP is essential for the inhibitory function of FcγRIIB, it also has critical modulating functions in signaling initiated from activating immunoreceptors such as B cell Ag receptor. In this study, we found that SHIP is indistinguishably recruited to the plasma membrane after BCR stimulation with or without FcγRIIB coligation in human cell lines and primary cells. Interestingly, fluorescence recovery after photobleaching analysis reveals differential mobility of SHIP-enhanced GFP depending on the mode of stimulation, suggesting that although BCR and FcγRIIB can both recruit SHIP, this occurs via distinct molecular complexes. Mutagenesis of a SHIP-enhanced GFP fusion protein reveals that the SHIP-Src homology 2 domain is essential in both cases whereas the C terminus is required for recruitment via BCR stimulation, but is less important with FcγRIIB coligation. Experiments with pharmacological inhibitors reveal that Syk activity is required for optimal stimulation-induced membrane localization of SHIP, whereas neither PI3K or Src kinase activity is essential. BCR-induced association of SHIP with binding partner Shc1 is dependent on Syk, as is tyrosine phosphorylation of both partners. Our results indicate that FcγRIIB is not uniquely able to promote membrane recruitment of SHIP, but rather modulates its function via formation of distinct signaling complexes. Membrane recruitment of SHIP via Syk-dependent mechanisms may be an important factor modulating immunoreceptor signaling.
Collapse
Affiliation(s)
- Samantha D Pauls
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada
| | - Arnab Ray
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada; and
| | - Sen Hou
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada; and
| | - Andrew T Vaughan
- Cancer Sciences Unit, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Mark S Cragg
- Cancer Sciences Unit, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Aaron J Marshall
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada; Department of Immunology, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada; and
| |
Collapse
|
21
|
Lutz J, Dittmann K, Bösl MR, Winkler TH, Wienands J, Engels N. Reactivation of IgG-switched memory B cells by BCR-intrinsic signal amplification promotes IgG antibody production. Nat Commun 2015; 6:8575. [PMID: 26815242 PMCID: PMC4633962 DOI: 10.1038/ncomms9575] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 09/05/2015] [Indexed: 01/26/2023] Open
Abstract
Secondary antibody responses are marked by faster kinetics, improved antibody affinity and a switch from IgM to other immunoglobulin isotypes, most notably IgG, compared with primary responses. These changes protect from reinfection and represent the principle of most vaccination strategies. Yet, the molecular mechanisms that underlie B-cell memory responses are unclear. Here we show, by inactivating the immunoglobulin tail tyrosine (ITT) signalling motif of membrane-bound IgG1 in the mouse, that the ITT facilitates maintenance and reactivation of IgG-switched memory B cells in vivo. The ITT motif equips IgG-switched cells with enhanced BCR signalling capacity, which supports their competitiveness in secondary immune reactions and drives the formation of IgG-secreting plasma cells even in the absence of T-cell help. Our results demonstrate that ITT signalling promotes the vigorous production of IgG antibodies and thus provide a molecular basis for humoral immunological memory.
Collapse
Affiliation(s)
- Johannes Lutz
- Institute of Cellular and Molecular Immunology, Georg-August-University of Göttingen, Medical Faculty, Humboldtallee 34, 37073 Göttingen, Germany
| | - Kai Dittmann
- Institute of Cellular and Molecular Immunology, Georg-August-University of Göttingen, Medical Faculty, Humboldtallee 34, 37073 Göttingen, Germany
| | - Michael R Bösl
- Max Planck Institute of Neurobiology, Transgenic Core Facility, 82152 Martinsried, Germany
| | - Thomas H Winkler
- Hematopoiesis Unit, Department of Biology, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Glückstrasse 6, 91054 Erlangen, Germany
| | - Jürgen Wienands
- Institute of Cellular and Molecular Immunology, Georg-August-University of Göttingen, Medical Faculty, Humboldtallee 34, 37073 Göttingen, Germany
| | - Niklas Engels
- Institute of Cellular and Molecular Immunology, Georg-August-University of Göttingen, Medical Faculty, Humboldtallee 34, 37073 Göttingen, Germany
| |
Collapse
|
22
|
Bretscher PA. A Conversation with Cohn on the Activation of CD4 T Cells. Scand J Immunol 2015; 82:147-59. [PMID: 25998043 PMCID: PMC4736666 DOI: 10.1111/sji.12315] [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] [Received: 12/09/2014] [Accepted: 04/17/2015] [Indexed: 01/07/2023]
Abstract
Despite an agreement on most issues surrounding models for how lymphocytes are activated and inactivated, and arising out of the 1970 Two Signal Model of lymphocyte activation, Cohn and I have different perspectives on two critical issues concerning the activation of CD4 T cells. One issue is the origin of the first effector T helper (eTh) cells, postulated by both of us to be required to optimally activate precursor Th (pTh), that is naïve CD4 T cells, to further generate eTh cells. The second issue arises from our agreement that the antigen‐dependent CD4 T cell cooperation, that we both postulate is required to activate naïve CD4 T (pTh) cells, most likely is mediated by the operational recognition of linked epitopes. Although agreeing on the centrality of this operational mechanism, we disagree about how it might be realized at the molecular/cellular level. I respond here to issues raised by Cohn concerning these two mechanistic questions, in his response to my recent article on the activation and inactivation of mature CD4 T cells.
Collapse
Affiliation(s)
- P A Bretscher
- Department of Microbiology and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
23
|
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
|
24
|
Wienands J, Engels N. The Memory Function of the B Cell Antigen Receptor. Curr Top Microbiol Immunol 2015; 393:107-121. [PMID: 26362935 DOI: 10.1007/82_2015_480] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Activated B lymphocytes preserve their antigen experience by differentiating into long-lived pools of antibody-secreting plasma cells or various types of memory B cells (MBCs). The former population constantly produces serum immunoglobulins with sufficient specificity and affinity to thwart infections with recurrent pathogens. By contrast, memory B cell populations retain their antigen receptors on the cell surface and hence need pathogen-induced differentiation steps before they can actively contribute to host defense. The terminal differentiation of MBCs into antibody-secreting plasma cells is hallmarked by the absence of the lag phase characteristic for primary antibody responses. Moreover, secondary antibody responses are predominantly driven by MBCs that bear an antigen receptor of the IgG class on their surface although IgM-positive memory populations exist as well. These fundamental principles of B cell memory were enigmatic for decades. Only recently, we have begun to understand the underlying mechanisms. This review summarizes our current understanding of how different subpopulations of MBCs are generated during primary immune responses and how their functional heterogeneity on antigen recall is controlled by different signaling capabilities of B cell antigen receptor (BCR) isotypes and by the nature of the antigen.
Collapse
Affiliation(s)
- Jürgen Wienands
- Medical Faculty, Institute of Cellular and Molecular Immunology, Georg-August-University of Göttingen, Humboldtallee 34, 37073, Göttingen, Germany.
| | - Niklas Engels
- Medical Faculty, Institute of Cellular and Molecular Immunology, Georg-August-University of Göttingen, Humboldtallee 34, 37073, Göttingen, Germany.
| |
Collapse
|
25
|
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: 75] [Impact Index Per Article: 6.8] [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
|
26
|
Goulabchand R, Vincent T, Batteux F, Eliaou JF, Guilpain P. Impact of autoantibody glycosylation in autoimmune diseases. Autoimmun Rev 2014; 13:742-50. [DOI: 10.1016/j.autrev.2014.02.005] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 02/01/2014] [Indexed: 12/12/2022]
|
27
|
Szili D, Cserhalmi M, Bankó Z, Nagy G, Szymkowski DE, Sármay G. Suppression of innate and adaptive B cell activation pathways by antibody coengagement of FcγRIIb and CD19. MAbs 2014; 6:991-9. [PMID: 24828435 DOI: 10.4161/mabs.28841] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The Fc receptor (FcγRIIb) inhibits B cell responses when coengaged with B cell receptor (BCR), and has become a target for new autoimmune disease therapeutics. For example, BCR and FcγRIIb coengagement via the Fc-engineered anti-CD19 XmAb5871 suppresses humoral immune responses. We now assess effects of XmAb5871 on other activation pathways, including the pathogen-associated molecular pattern receptor, TLR9. Since TLR9 signaling is implicated in autoimmune diseases, we asked if XmAb5871 could inhibit TLR9 costimulation. We show that XmAb5871 decreases ERK and AKT activation, cell proliferation, cytokine, and IgG production induced by BCR and/or TLR9 signals. XmAb5871 also inhibited differentiation of citrullinated peptide-specific plasma cells from rheumatoid arthritis patients. XmAb5871 may therefore have potential to suppress pathogenic B cells in autoimmune diseases.
Collapse
Affiliation(s)
- Dániel Szili
- Department of Immunology; Eötvös Loránd University; Budapest, Hungary
| | - Marcell Cserhalmi
- Department of Immunology; Eötvös Loránd University; Budapest, Hungary
| | - Zsuzsanna Bankó
- Department of Immunology; Eötvös Loránd University; Budapest, Hungary
| | - György Nagy
- Department of Rheumatology; Polyclinic of the Hospitaller Brothers of St. John of God; Budapest, Hungary
| | | | | |
Collapse
|
28
|
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
|
29
|
Mimoto F, Katada H, Kadono S, Igawa T, Kuramochi T, Muraoka M, Wada Y, Haraya K, Miyazaki T, Hattori K. Engineered antibody Fc variant with selectively enhanced FcγRIIb binding over both FcγRIIa(R131) and FcγRIIa(H131). Protein Eng Des Sel 2013; 26:589-98. [PMID: 23744091 PMCID: PMC3785249 DOI: 10.1093/protein/gzt022] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 04/24/2013] [Accepted: 05/08/2013] [Indexed: 12/02/2022] Open
Abstract
Engaging inhibitory FcγRIIb by Fc region has been recently reported to be an attractive approach for improving the efficacy of antibody therapeutics. However, the previously reported S267E/L328F variant with enhanced binding affinity to FcγRIIb, also enhances binding affinity to FcγRIIa(R131) allotype to a similar degree because FcγRIIb and FcγRIIa(R131) are structurally similar. In this study, we applied comprehensive mutagenesis and structure-guided design based on the crystal structure of the Fc/FcγRIIb complex to identify a novel Fc variant with selectively enhanced FcγRIIb binding over both FcγRIIa(R131) and FcγRIIa(H131). This novel variant has more than 200-fold stronger binding affinity to FcγRIIb than wild-type IgG1, while binding affinity to FcγRIIa(R131) and FcγRIIa(H131) is comparable with or lower than wild-type IgG1. This selectivity was achieved by conformational change of the C(H)2 domain by mutating Pro to Asp at position 238. Fc variant with increased binding to both FcγRIIb and FcγRIIa induced platelet aggregation and activation in an immune complex form in vitro while our novel variant did not. When applied to agonistic anti-CD137 IgG1 antibody, our variant greatly enhanced the agonistic activity. Thus, the selective enhancement of FcγRIIb binding achieved by our Fc variant provides a novel tool for improving the efficacy of antibody therapeutics.
Collapse
Affiliation(s)
| | | | | | - T. Igawa
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
With the approval by the FDA in 2011 of a biologic agent (namely belimumab) for the treatment of systemic lupus erythematosus (SLE), optimism abounds that additional biologic (and nonbiologic) agents will be similarly endorsed. Given the numerous immune-based abnormalities associated with SLE, the potential therapeutic targets for biologic agents and the candidate biologic approaches are also numerous. These approaches include: biologic agents that promote B-cell depletion, B-cell inactivation, or the generation of regulatory B cells; biologic agents that induce T-cell tolerance, block T-cell activation and differentiation, or alter T-cell trafficking; biologic agents that target the B-cell activating factor (BAFF) axis, type I interferons, IL-6 and its receptor, or TNF; and the adoptive transfer of ex vivo-generated regulatory T cells. Owing to the great heterogeneity inherent to SLE, no single approach should be expected to be effective in all patients. As our understanding of the pathogenic mechanisms of SLE continues to expand, additional therapeutic targets and approaches will undoubtedly be identified and should be fully exploited.
Collapse
|
31
|
Hess C, Winkler A, Lorenz AK, Holecska V, Blanchard V, Eiglmeier S, Schoen AL, Bitterling J, Stoehr AD, Petzold D, Schommartz T, Mertes MMM, Schoen CT, Tiburzy B, Herrmann A, Köhl J, Manz RA, Madaio MP, Berger M, Wardemann H, Ehlers M. T cell-independent B cell activation induces immunosuppressive sialylated IgG antibodies. J Clin Invest 2013; 123:3788-96. [PMID: 23979161 DOI: 10.1172/jci65938] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 06/27/2013] [Indexed: 01/06/2023] Open
Abstract
Antigen-specific Abs are able to enhance or suppress immune responses depending on the receptors that they bind on immune cells. Recent studies have shown that pro- or antiinflammatory effector functions of IgG Abs are also regulated through their Fc N-linked glycosylation patterns. IgG Abs that are agalactosylated (non-galactosylated) and asialylated are proinflammatory and induced by the combination of T cell-dependent (TD) protein antigens and proinflammatory costimulation. Sialylated IgG Abs, which are immunosuppressive, and Tregs are produced in the presence of TD antigens under tolerance conditions. T cell-independent (TI) B cell activation via B cell receptor (BCR) crosslinking through polysaccharides or via BCR and TLR costimulation also induces IgG Abs, but the Fc glycosylation state of these Abs is unknown. We found in mouse experiments that TI immune responses induced suppressive sialylated IgGs, in contrast to TD proinflammatory Th1 and Th17 immune responses, which induced agalactosylated and asialylated IgGs. Transfer of low amounts of antigen-specific sialylated IgG Abs was sufficient to inhibit B cell activation and pathogenic immune reactions. These findings suggest an immune regulatory function for TI immune responses through the generation of immunosuppressive sialylated IgGs and may provide insight on the role of TI immune responses during infection, vaccination, and autoimmunity.
Collapse
Affiliation(s)
- Constanze Hess
- Laboratory of Tolerance and Autoimmunity, German Rheumatism Research Center, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Montaudouin C, Anson M, Hao Y, Duncker SV, Fernandez T, Gaudin E, Ehrenstein M, Kerr WG, Colle JH, Bruhns P, Daëron M, Freitas AA. Quorum sensing contributes to activated IgM-secreting B cell homeostasis. THE JOURNAL OF IMMUNOLOGY 2012; 190:106-14. [PMID: 23209322 DOI: 10.4049/jimmunol.1200907] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Maintenance of plasma IgM levels is critical for immune system function and homeostasis in humans and mice. However, the mechanisms that control homeostasis of the activated IgM-secreting B cells are unknown. After adoptive transfer into immune-deficient hosts, B lymphocytes expand poorly, but fully reconstitute the pool of natural IgM-secreting cells and circulating IgM levels. By using sequential cell transfers and B cell populations from several mutant mice, we were able to identify novel mechanisms regulating the size of the IgM-secreting B cell pool. Contrary to previous mechanisms described regulating homeostasis, which involve competition for the same niche by cells having overlapping survival requirements, homeostasis of the innate IgM-secreting B cell pool is also achieved when B cell populations are able to monitor the number of activated B cells by detecting their secreted products. Notably, B cell populations are able to assess the density of activated B cells by sensing their secreted IgG. This process involves the FcγRIIB, a low-affinity IgG receptor that is expressed on B cells and acts as a negative regulator of B cell activation, and its intracellular effector the inositol phosphatase SHIP. As a result of the engagement of this inhibitory pathway, the number of activated IgM-secreting B cells is kept under control. We hypothesize that malfunction of this quorum-sensing mechanism may lead to uncontrolled B cell activation and autoimmunity.
Collapse
Affiliation(s)
- Caroline Montaudouin
- Unité de Biologie des Populations Lymphocytaires, Départment d'Immunologie, Institut Pasteur, Paris 75015, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Olovnikova NI, Ershler MA, Grigorieva OV, Petrov AV, Miterev GY. Impact on N-glycosylation profile of monoclonal anti-D antibodies as a way to control their immunoregulatory and cytotoxic properties. BIOCHEMISTRY (MOSCOW) 2012; 77:925-33. [PMID: 22860915 DOI: 10.1134/s0006297912080147] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Prophylaxis of hemolytic disease of newborns is based on the ability of polyclonal anti-D antibodies for suppressing maternal immune response against D-positive fetal red blood cells. The immunosuppressive effect of anti-D antibody is mediated by interaction between its Fc-fragment and low-affinity IgG Fc-receptor (FcγR) on the immune cell. No clinically effective monoclonal anti-D antibody (mAb) that can replace polyclonal anti-D immunoglobulin has been developed yet. The goals of this study were comparison of structural and functional properties of human anti-D polyclonal and monoclonal Abs and assessment of the possibility to manipulate the effector properties of the mAb. N-Glycosylation and particularly the content of nonfucosylated glycans are crucial for affinity of mAb to FcγRIIIA, which plays the key role in the clearance of sensitized cells. We studied and compared glycoprofiles and FcγRIIIA-mediated hemolytic ability of human polyclonal antibodies and anti-D mAbs produced by human B-cell lines, human-rodent heterohybridomas, and a human non-lymphoid cell line PER.C6. Replacement of producing cell line and use of glycosylation modulators can convert an inert mAb into an active one. Nevertheless, rodent cell lines, as well as human non-lymphoid cells, distort natural glycosylation of human IgG and could lead to the loss of immunosuppressive properties. All of the anti-D mAbs secreted by human B-cell lines have a glycoprofile close to human serum IgG. Hence, the constant ratio of IgG glycoforms in human serum is predetermined by glycosylation at the level of the individual antibody-producing cell. The anti-D fraction of polyclonal anti-D immunoglobulin compared to the total human IgG contains more nonfucosylated glycans. Thus, only human transformed B-cells are an appropriate source for efficient anti-D mAbs that can imitate the action of polyclonal anti-D IgG.
Collapse
Affiliation(s)
- N I Olovnikova
- Hematology Research Center, Ministry of Health and Social Development, Noviy Zykovskiy Proezd 4A, 125167 Moscow, Russia.
| | | | | | | | | |
Collapse
|
34
|
Baseline polysaccharide-specific antibodies may not consistently inhibit booster antibody responses in infants to a serogroup C meningococcal protein–polysaccharide conjugate vaccine. Vaccine 2012; 30:4153-9. [DOI: 10.1016/j.vaccine.2012.04.063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Revised: 04/12/2012] [Accepted: 04/19/2012] [Indexed: 11/18/2022]
|
35
|
Reynaud CA, Descatoire M, Dogan I, Huetz F, Weller S, Weill JC. IgM memory B cells: a mouse/human paradox. Cell Mol Life Sci 2012; 69:1625-34. [PMID: 22481437 PMCID: PMC3337997 DOI: 10.1007/s00018-012-0971-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Revised: 03/13/2012] [Accepted: 03/13/2012] [Indexed: 11/14/2022]
Abstract
Humoral memory is maintained by two types of persistent cells, memory B cells and plasma cells, which have different phenotypes and functions. Long-lived plasma cells can survive for a lifespan within a complex niche in the bone marrow and provide continuous protective serum antibody levels. Memory B cells reside in secondary lymphoid organs, where they can be rapidly mobilized upon a new antigenic encounter. Surface IgG has long been taken as a surrogate marker for memory in the mouse. Recently, however, we have brought evidence for a long-lived IgM memory B cell population in the mouse, while we have also argued that, in humans, these same cells are not classical memory B cells but marginal zone (MZ) B cells which, as opposed to their mouse MZ counterpart, recirculate and carry a mutated B cell receptor. In this review, we will discuss these apparently paradoxical results.
Collapse
Affiliation(s)
- Claude-Agnès Reynaud
- Faculté de Médecine, Site Necker-Enfants Malades, INSERM U783 “Développement du système immunitaire”, Université Paris Descartes, 156 rue de Vaugirard, 75730 Paris Cedex 15, France
| | - Marc Descatoire
- Faculté de Médecine, Site Necker-Enfants Malades, INSERM U783 “Développement du système immunitaire”, Université Paris Descartes, 156 rue de Vaugirard, 75730 Paris Cedex 15, France
| | - Ismail Dogan
- Faculté de Médecine, Site Necker-Enfants Malades, INSERM U783 “Développement du système immunitaire”, Université Paris Descartes, 156 rue de Vaugirard, 75730 Paris Cedex 15, France
| | - François Huetz
- Faculté de Médecine, Site Necker-Enfants Malades, INSERM U783 “Développement du système immunitaire”, Université Paris Descartes, 156 rue de Vaugirard, 75730 Paris Cedex 15, France
- Unité de Biologie des Populations Lymphocytaires, CNRS URA 1961, Institut Pasteur, 25 rue du Docteur Roux, 75724 Paris Cedex 15, France
| | - Sandra Weller
- Faculté de Médecine, Site Necker-Enfants Malades, INSERM U783 “Développement du système immunitaire”, Université Paris Descartes, 156 rue de Vaugirard, 75730 Paris Cedex 15, France
| | - Jean-Claude Weill
- Faculté de Médecine, Site Necker-Enfants Malades, INSERM U783 “Développement du système immunitaire”, Université Paris Descartes, 156 rue de Vaugirard, 75730 Paris Cedex 15, France
| |
Collapse
|
36
|
Amezcua Vesely MC, Schwartz M, Bermejo DA, Montes CL, Cautivo KM, Kalergis AM, Rawlings DJ, Acosta-Rodríguez EV, Gruppi A. FcγRIIb and BAFF differentially regulate peritoneal B1 cell survival. THE JOURNAL OF IMMUNOLOGY 2012; 188:4792-800. [PMID: 22516957 DOI: 10.4049/jimmunol.1102070] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
B1 cells produce most natural Abs in unimmunized mice and play a key role in the response to thymus-independent Ags and microbial infection. Enlargement of B1 cell number in mice is often associated with autoimmunity. However, the factors that control peripheral B1 cell survival remain poorly characterized. Mice lacking the inhibitory receptor FcγRIIb exhibit a massive expansion in peritoneal B1 cells, implicating this receptor in B1 cell homeostasis. In this study, we show that peritoneal B1 cells express the highest levels of FcγRIIb among B cell subsets and are highly susceptible to FcγRIIb-mediated apoptosis. B1 cells upregulate FcγRIIb in response to innate signals, including CpG, and the B cell homeostatic cytokine BAFF efficiently protects activated B1 cells from FcγRIIb-mediated apoptosis via receptor downregulation. BAFF-transgenic mice manifest an expansion of peritoneal B1 cells that express lower levels of FcγRIIb and exhibit reduced susceptibility to apoptosis. Whereas both peritoneal B1 cells from wild-type and BAFF-transgenic mice immunized with CpG exhibit an increase in FcγRIIb levels, this change is blunted in BAFF-transgenic animals. Our combined results demonstrate that FcγRIIb controls peritoneal B1 cell survival and this program can be modulated by the BAFF signaling axis.
Collapse
Affiliation(s)
- María C Amezcua Vesely
- Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba CP 5000, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Chu SY, Horton HM, Pong E, Leung IWL, Chen H, Nguyen DH, Bautista C, Muchhal US, Bernett MJ, Moore GL, Szymkowski DE, Desjarlais JR. Reduction of total IgE by targeted coengagement of IgE B-cell receptor and FcγRIIb with Fc-engineered antibody. J Allergy Clin Immunol 2012; 129:1102-15. [PMID: 22257644 DOI: 10.1016/j.jaci.2011.11.029] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 11/16/2011] [Accepted: 11/18/2011] [Indexed: 01/16/2023]
Abstract
BACKGROUND Sequestration of IgE to prevent its binding to high-affinity IgE receptor FcεRI on basophils and mast cells is an effective therapy for allergic asthma. IgE production requires differentiation of activated IgE(+) B cells into plasma cells upon allergen sensitization. B-cell receptor signaling is suppressed by the inhibitory IgG Fc receptor FcγRIIb; therefore, we reasoned that a therapeutic antibody that coengages FcγRIIb and IgE B-cell receptor would not only sequester IgE but also suppress its production by blocking IgE(+) B-cell activation and differentiation to IgE-secreting plasma cells. OBJECTIVE To explore the effects of IgE sequestration versus IgE suppression by comparing omalizumab to FcγRIIb-optimized anti-IgE antibodies in humanized mouse models of immunoglobulin production. METHODS By using a murine anti-IgE antibody as a template, we humanized, increased IgE binding, and modified its Fc domain to increase affinity for FcγRIIb. We next compared effects of this antibody (XmAb7195) versus omalizumab on the secretion of IgE and other isotypes in human PBMC cultures and in PBMC-engrafted severe combined immunodeficiency mice. RESULTS Relative to omalizumab, XmAb7195 has a 5-fold higher affinity for human IgE and more than 400-fold higher affinity for FcγRIIb. In addition to sequestering soluble IgE, XmAb7195 inhibited plasma cell differentiation and consequent human IgE production through coengagement of IgE B-cell receptor with FcγRIIb. In PBMC-engrafted mice, XmAb7195 reduced total human IgE (but not IgG or IgM) levels by up to 40-fold relative to omalizumab. CONCLUSION XmAb7195 acts by IgE sequestration coupled with an FcγRIIb-mediated inhibitory mechanism to suppress the formation of IgE-secreting plasma cells and reduce both free and total IgE levels.
Collapse
|
38
|
Modulation of the humoral immune response by targeting CD40 and FcγRII/III; delivery of soluble but not particulate antigen to CD40 enhances antibody responses with a Th1 bias. Mol Immunol 2011; 49:155-62. [DOI: 10.1016/j.molimm.2011.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 08/08/2011] [Accepted: 08/09/2011] [Indexed: 12/25/2022]
|
39
|
Zhao XQ, Zhang MW, Wang F, Zhao YX, Li JJ, Wang XP, Bu PL, Yang JM, Liu XL, Zhang MX, Gao F, Zhang C, Zhang Y. CRP enhances soluble LOX-1 release from macrophages by activating TNF-α converting enzyme. J Lipid Res 2011; 52:923-33. [PMID: 21364202 DOI: 10.1194/jlr.m015156] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Circulating levels of soluble lectin-like oxidized low-density lipoprotein receptor-1 (sLOX-1) play an important role in the development and progression of atherosclerosis. We hypothesized that the inflammatory marker C-reactive protein (CRP) might stimulate sLOX-1 release by activating tumor necrosis factor-α converting enzyme (TACE). Macrophages differentiated from THP-1 cells were stimulated with TNF-α and further treated with CRP in the absence or presence of specific inhibitors or small interfering RNA (siRNA). Our results showed that CRP increased sLOX-1 release from activated macrophages in a dose-dependent manner and that these effects were regulated by Fc γ receptor II (FcγRII)-mediated p47(phox) phosphorylation, reactive oxygen species (ROS) production, and TACE activation. CRP also enhanced sLOX-1 release from macrophages derived from peripheral blood mononuclear cells (PBMC) of patients with acute coronary syndrome (ACS). Pretreatment with antibody against FcγRII or with CD32 siRNA, p47(phox) siRNA, apocynin, N-acetylcysteine, tumor necrosis factor-α protease inhibitor 1 (TAPI-1) or TACE siRNA attenuated sLOX-1 release induced by CRP. CRP also elevated serum sLOX-1 levels in a rabbit model of atherosclerosis. Thus, CRP might stimulate sLOX-1 release, and the underlying mechanisms possibly involved FcγRII-mediated p47(phox) phosphorylation, ROS production, and TACE activation.
Collapse
Affiliation(s)
- Xue Qiang Zhao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Horton HM, Chu SY, Ortiz EC, Pong E, Cemerski S, Leung IWL, Jacob N, Zalevsky J, Desjarlais JR, Stohl W, Szymkowski DE. Antibody-Mediated Coengagement of FcγRIIb and B Cell Receptor Complex Suppresses Humoral Immunity in Systemic Lupus Erythematosus. THE JOURNAL OF IMMUNOLOGY 2011; 186:4223-33. [DOI: 10.4049/jimmunol.1003412] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
41
|
Paradoxical suppression of poly-specific broadly neutralizing antibodies in the presence of strain-specific neutralizing antibodies following HIV infection. J Theor Biol 2011; 277:55-66. [PMID: 21315731 DOI: 10.1016/j.jtbi.2011.01.050] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 01/17/2011] [Accepted: 01/31/2011] [Indexed: 11/21/2022]
Abstract
One of the first immunologic responses against HIV infection is the presence of neutralizing antibodies that seem able to inactivate several HIV strains. Moreover, in vitro studies have shown the existence of monoclonal antibodies that exhibit broad crossclade neutralizing potential. Yet their number is low and slow to develop in vivo. In this paper, we investigate the potential benefits of inducing poly-specific neutralizing antibodies in vivo throughout immunization. We develop a mathematical model that considers the activation of families of B lymphocytes producing poly-specific and strain-specific antibodies and use it to demonstrate that, even if such families are successful in producing neutralizing antibodies, the competition between them may limit the poly-specific response allowing the virus to escape. We modify this model to account for viral evolution under the pressure of antibody responses in natural HIV infection. The model can reproduce viral escape under certain conditions of B lymphocyte competition. Using these models we provide explanations for the observed antibody failure in controlling natural infection and predict quantitative measures that need to be satisfied for long-term control of HIV infection.
Collapse
|
42
|
Deficiency of activating Fcγ-receptors reduces hepatic clearance and deposition of IC and increases CIC levels in mercury-induced autoimmunity. PLoS One 2010; 5:e13413. [PMID: 20976163 PMCID: PMC2955531 DOI: 10.1371/journal.pone.0013413] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2010] [Accepted: 08/31/2010] [Indexed: 12/02/2022] Open
Abstract
Background Inorganic mercury (Hg) induces a T-cell dependent, systemic autoimmune condition (HgIA) where activating Fcγ-receptors (FcγRs) are important for the induction. In this study we examined the influence of activating FcγRs on circulating levels and organ localization of immune complexes (IC) in HgIA. Methods and Principal Findings Mercury treated BALB/c wt mice showed a significant but modest increase of circulating IC (CIC) from day 12 until day 18 and day 35 for IgG2a- and IgG1- CIC, respectively. Mercury-treated mice lacking the trans-membrane γ-chain of activating FcγRs (FcRγ−/−) had significantly higher CIC levels of both IgG1-CIC and IgG2a-CIC than wt mice during the treatment course. The hepatic uptake of preformed CIC was significantly more efficient in wt mice compared to FcγR−/− mice, but also development of extrahepatic tissue IC deposits was delayed in FcRγ−/− mice. After 35 days of Hg treatment the proportion of immune deposits, as well as the amounts was significantly reduced in vessel FcRγ−/− mice compared to wt mice. Conclusions We conclude that mice lacking functional activating FcγRs respond to Hg with increased levels and altered quality of CIC compared with wt mice. Lack of functional activating FcγRs delayed the elimination of CIC, but also significantly reduced extrahepatic tissue localization of CIC.
Collapse
|
43
|
Fang Y, Larsson L, Mattsson J, Lycke N, Xiang Z. Mast cells contribute to the mucosal adjuvant effect of CTA1-DD after IgG-complex formation. THE JOURNAL OF IMMUNOLOGY 2010; 185:2935-41. [PMID: 20675596 DOI: 10.4049/jimmunol.1000589] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mast cell activation is one of the most dramatic immune-mediated responses the body can encounter. In the worst scenario (i.e., anaphylaxis), this response is fatal. However, the importance of mast cells as initiators and effectors of both innate and adaptive immunity in healthy individuals has recently been appreciated. It was reported that mast cell activation can be used as an adjuvant to promote Ag-specific humoral immune responses upon vaccination. In this study, we have used a clinically relevant mucosal adjuvant, cholera toxin A1 subunit (CTA1)-DD, which is a fusion protein composed of CTA1, the ADP-ribosylating part of cholera toxin, and DD, two Ig-binding domains derived from Staphylococcus aureus protein A. CTA1-DD in combination with polyclonal IgG induced degranulation and production of TNF-alpha from mouse mast cells. Furthermore, CTA1-DD and polyclonal IgG complex induced mast cell degranulation in mouse skin tissue and nasal mucosa. We also found that intranasal immunization with hapten (4-hydroxy-3-nitrophenyl) acetyl (NP) coupled to chicken gammaglobulin admixed with CTA1-DD complexed with polyclonal IgG greatly enhanced serum IgG anti-NP Ab responses and stimulated higher numbers of NP-specific plasma cells in the bone marrow as compared with that observed in mice immunized with NP-chicken gammaglobulin with CTA1-DD alone. This CTA1-DD/IgG complex-mediated enhancement was mast cell dependent because it was absent in mast cell-deficient Kit(W-sh/W-sh) mice. In conclusion, our data suggest that a clinically relevant adjuvant, CTA1-DD, exerts additional augmenting effects through activation of mucosal mast cells, clearly demonstrating that mast cells could be further exploited for improving the efficacy of mucosal vaccines.
Collapse
Affiliation(s)
- Yu Fang
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Research Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | |
Collapse
|
44
|
Victor JR, Muniz BP, Fusaro AE, de Brito CA, Taniguchi EF, Duarte AJS, Sato MN. Maternal immunization with ovalbumin prevents neonatal allergy development and up-regulates inhibitory receptor Fc gamma RIIB expression on B cells. BMC Immunol 2010; 11:11. [PMID: 20222978 PMCID: PMC2848204 DOI: 10.1186/1471-2172-11-11] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Accepted: 03/11/2010] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Preconception allergen immunization prevents neonatal allergen sensitization in mice by a complex interaction between regulatory cells/factors and antibodies. The present study assessed the influence of maternal immunization with ovalbumin (OVA) on the immune response of 3 day-old and 3 week-old offspring immunized or non-immunized with OVA and evaluated the effect of IgG treatment during fetal development or neonatal period. RESULTS Maternal immunization with OVA showed increased levels of Fc gamma RIIb expression in splenic B cells of neonates, which were maintained for up to 3 weeks and not affected by additional postnatal OVA immunization. Maternal immunization also exerted a down-modulatory effect on both IL-4 and IFN-gamma-secreting T cells and IL-4 and IL-12- secreting B cells. Furthermore, immunized neonates from immunized mothers showed a marked inhibition of antigen-specific IgE Ab production and lowered Th2/Th1 cytokine levels, whereas displaying enhanced Fc gamma RIIb expression on B cells. These offspring also showed reduced antigen-specific proliferative response and lowered B cell responsiveness. Moreover, in vitro evaluation revealed an impairment of B cell activation upon engagement of B cell antigen receptor by IgG from OVA-immunized mice. Finally, in vivo IgG transference during pregnancy or breastfeeding revealed that maternal Ab transference was able to increase regulatory cytokines, such as IL-10, in the prenatal stage; yet only the postnatal treatment prevented neonatal sensitization. None of the IgG treatments induced immunological changes in the offspring, as it was observed for those from OVA-immunized mothers. CONCLUSION Maternal immunization upregulates the inhibitory Fc gamma RIIb expression on offspring B cells, avoiding skewed Th2 response and development of allergy. These findings contribute to the advancement of prophylactic strategies to prevent allergic diseases in early life.
Collapse
Affiliation(s)
- Jefferson R Victor
- Laboratory of Dermatology and Immunodeficiencies, School of Medicine, University of São Paulo, LIM 56, São Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
45
|
Angyal A, Szekeres Z, Balogh P, Neer Z, Szarka E, Virag V, Medgyesi D, Prechl J, Sarmay G. CD16/32-specific biotinylated 2.4G2 single-chain Fv complexed with avidin-FITC enhances FITC-specific humoral immune response in vivo in a CD16-dependent manner. Int Immunol 2009; 22:71-80. [PMID: 19951957 DOI: 10.1093/intimm/dxp115] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Fcgamma receptors (FcgammaRs) play an essential role in the regulation of immune response due to their ability to bind immune complexes. Activating FcgammaRs may facilitate antigen presentation and dendritic-cell maturation, while in the late phase of the immune response, the inhibitory FcgammaRIIb may down-regulate B-cell activation upon cross-linking with activating receptors. In this study, we investigated the in vivo role of FcgammaRs on the modulation of humoral immune response. In order to get well-defined immune complexes that can bind to both the activating and the inhibitory FcgammaRs, we designed a mono-biotinylated single-chain fragment variable construct from the rat anti-mouse CD16/32 clone 2.4G2, linked to avidin-FITC, and tested its effect on the FITC-hapten-specific T-independent type 2 (TI-2) and T-dependent (TD) immune response. When injected intravenously in mice, the complex bound to a small portion of B220+, CD11b(high) and CD11c(high) cells and was localized in the spleen on marginal zone macrophages 15 min after treatment. When applied as a booster following primary immunization with TI-2 (FITC-dextran) or TD (FITC-keyhole limpet haemocyanin) antigens, the complex elevated the number of hapten-specific IgM/IgG-producing B cells. This effect was diminished in CD16KO mice, suggesting that the activating-type FcgammaRIII might be a key mediator of this mechanism.
Collapse
Affiliation(s)
- Adrienn Angyal
- Department of Immunology, Eotvos Lorand University, 1117 Budapest, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Robinette RA, Oli MW, McArthur WP, Brady LJ. Beneficial immunomodulation by Streptococcus mutans anti-P1 monoclonal antibodies is Fc independent and correlates with increased exposure of a relevant target epitope. THE JOURNAL OF IMMUNOLOGY 2009; 183:4628-38. [PMID: 19752237 DOI: 10.4049/jimmunol.0803300] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We showed previously that deliberate immunization of BALB/c mice with immune complexes (IC) of the cariogenic bacterium Streptococcus mutans and mAbs against its surface adhesin P1 results in changes in the specificity and isotype of elicited anti-P1 Abs. Depending on the mAb, changes were beneficial, neutral, or detrimental, as measured by the ability of the serum from immunized mice to inhibit bacterial adherence to human salivary agglutinin by a BIAcore surface plasmon resonance assay. The current study further defined changes in the host response that result from immunization with IC containing beneficial mAbs, and evaluated mechanisms by which beneficial immunomodulation could occur in this system. Immunomodulatory effects varied depending upon genetic background, with differing results in C57BL/6 and BALB/c mice. Desirable effects following IC immunization were observed in the absence of activating FcRs in BALB/c Fcer1g transgenic mice. mAb F(ab')(2) mediated desirable changes similar to those observed using intact IgG. Sera from IC-immunized BALB/c mice that were better able to inhibit bacterial adherence demonstrated an increase in Abs able to compete with an adherence-inhibiting anti-P1 mAb, and binding of a beneficial immumomodulatory mAb to S. mutans increased exposure of that epitope. Consistent with a mechanism involving a mAb-mediated structural alteration of P1 on the cell surface, immunization with truncated P1 derivatives lacking segments that contribute to recognition by beneficial immunomodulatory mAbs resulted in an improvement in the ability of elicited serum Abs to inhibit bacterial adherence compared with immunization with the full-length protein.
Collapse
Affiliation(s)
- Rebekah A Robinette
- Department of Oral Biology, University of Florida College of Dentistry, P.O. Box 100424, Gainesville, FL 32610, USA.
| | | | | | | |
Collapse
|
47
|
Tallmadge RL, McLaughlin K, Secor E, Ruano D, Matychak MB, Flaminio MJBF. Expression of essential B cell genes and immunoglobulin isotypes suggests active development and gene recombination during equine gestation. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2009; 33:1027-1038. [PMID: 19442687 DOI: 10.1016/j.dci.2009.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2008] [Revised: 04/30/2009] [Accepted: 05/03/2009] [Indexed: 05/27/2023]
Abstract
Many features of the equine immune system develop during fetal life, yet the naïve or immature immune state of the neonate renders the foal uniquely susceptible to particular pathogens. RT-PCR and immunohistochemical experiments investigated the progressive expression of developmental B cell markers and immunoglobulins in lymphoid tissues from equine fetus, pre-suckle neonate, foal, and adult horses. Serum IgM, IgG isotype, and IgA concentrations were also quantified in pre-suckle foals and adult horses. The expression of essential B cell genes suggests active development and gene recombination during equine gestation, including immunoglobulin isotype switching. The corresponding production of IgM and IgG proteins is detectable in a limited scale at birth. Although the equine neonate humoral response seems competent, B cell activation factors derived from antigen presenting cells and T cells may control critical developmental regulation and immunoglobulin production during the initial months of life.
Collapse
Affiliation(s)
- Rebecca L Tallmadge
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | | | |
Collapse
|
48
|
Ruseva M, Kolev M, Dagnaes-Hansen F, Hansen SB, Takahashi K, Ezekowitz A, Thiel S, Jensenius JC, Gadjeva M. Mannan-binding lectin deficiency modulates the humoral immune response dependent on the genetic environment. Immunology 2009; 127:279-88. [PMID: 19476514 DOI: 10.1111/j.1365-2567.2008.03016.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Mannan-binding lectin (MBL) is a plasma protein implicated in innate immune defence against a broad range of microorganisms, including viruses. It is also thought that MBL plays a role in the recruitment of the specific clonal immune response. This was studied by injecting soluble hepatitis B surface antigen (HBsAg) intravenously into mice deficient in both MBL-A and MBL-C (MBL DKO mice). The MBL DKO animals on mixed genetic background (SV129EvSv x C57BL/6) produced higher antibody titres than the wild-type littermates. After primary challenge with the antigen the immunoglobulin M anti-HBsAg antibody titres were threefold higher in the MBL DKO mice than in the wild-type mice. Following the boost, the immunoglobulin G anti-HBsAg antibody titres were 10-fold higher in the MBL DKO mice, suggesting that MBL plays a role in a negative feedback regulation of adaptive immunity. However, the modulating effect of MBL was dependent on the genetic environment. The MBL DKO mice backcrossed on a C57BL/6 background showed the opposite response with the MBL DKO mice now producing fewer antibodies than the wild-type animals, whereas MBL deficiency in mice with the SV129EvSv background did not show any effect in antibody production. These findings indicate that the modifying effect of MBL on the humoral immune response is influenced by the genetic environment.
Collapse
Affiliation(s)
- Marieta Ruseva
- Department of Medical Microbiology and Immunology, University of Aarhus, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Preissler MT, Kaiser L, Drake JR, Gosselin EJ. Low-Level Signaling Generated by FcγRIIB-B Cell Receptor Co-Ligation Establishes a State of Global B Cell Receptor Nonresponsiveness. Immunol Invest 2009. [DOI: 10.1081/imm-47385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
50
|
Chattha KS, Firth MA, Hodgins DC, Shewen PE. Age related variation in expression of CD21 and CD32 on bovine lymphocytes: A cross-sectional study. Vet Immunol Immunopathol 2009; 130:70-8. [DOI: 10.1016/j.vetimm.2009.01.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 01/19/2009] [Accepted: 01/26/2009] [Indexed: 12/31/2022]
|