1
|
Heterogeneity and Functions of Tumor-Infiltrating Antibody Secreting Cells: Lessons from Breast, Ovarian, and Other Solid Cancers. Cancers (Basel) 2022; 14:cancers14194800. [PMID: 36230721 PMCID: PMC9563085 DOI: 10.3390/cancers14194800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary B cells are gaining increasing recognition as important contributors to the tumor microenvironment, influencing, positively or negatively, tumor growth, patient survival, and response to therapies. Antibody secreting cells (ASCs) constitute a variable fraction of tumor-infiltrating B cells in most solid tumors, and they produce tumor-specific antibodies that can drive distinct immune responses depending on their isotypes and specificities. In this review, we discuss the current knowledge of the heterogeneity of ASCs infiltrating solid tumors and how both their canonical and noncanonical functions shape antitumor immunity, with a special emphasis on breast and ovarian cancers. Abstract Neglected for a long time in cancer, B cells and ASCs have recently emerged as critical actors in the tumor microenvironment, with important roles in shaping the antitumor immune response. ASCs indeed exert a major influence on tumor growth, patient survival, and response to therapies. The mechanisms underlying their pro- vs. anti-tumor roles are beginning to be elucidated, revealing the contributions of their secreted antibodies as well as of their emerging noncanonical functions. Here, concentrating mostly on ovarian and breast cancers, we summarize the current knowledge on the heterogeneity of tumor-infiltrating ASCs, we discuss their possible local or systemic origin in relation to their immunoglobulin repertoire, and we review the different mechanisms by which antibody (Ab) subclasses and isoforms differentially impact tumor cells and anti-tumor immunity. We also discuss the emerging roles of cytokines and other immune modulators produced by ASCs in cancer. Finally, we propose strategies to manipulate the tumor ASC compartment to improve cancer therapies.
Collapse
|
2
|
Arnoult C, Brachet G, Cadena Castaneda D, Azzopardi N, Passot C, Desvignes C, Paintaud G, Heuzé-Vourc'h N, Watier H, Gouilleux-Gruart V. Crucial Role for Immune Complexes but Not FcRn in Immunization against Anti-TNF-α Antibodies after a Single Injection in Mice. THE JOURNAL OF IMMUNOLOGY 2017; 199:418-424. [PMID: 28584008 DOI: 10.4049/jimmunol.1601246] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 05/06/2017] [Indexed: 01/08/2023]
Abstract
The immunogenicity of infliximab and adalimumab is a major concern because patients may develop Abs also called antidrug Abs (ADA), directed against these anti-TNF-α Abs after just a few weeks of treatment. These ADAs can lead to a decrease in biologic concentration, which is associated with lower treatment efficacy. Our aim was to study the involvement of immune complexes and neonatal Fc receptor (FcRn) in the emergence of ADAs in the case of anti-TNF-α Abs. Wild type and FcRn knockout mice were injected once with either infliximab or adalimumab, alone or preincubated with TNF-α. Adalimumab cross-reacts with murine TNF-α whereas infliximab is species specific. When injected alone, only adalimumab elicited a humoral response. By preforming immune complexes with TNF-α, an anti-infliximab response was elicited. Surprisingly, both wild type and FcRn knockout mice were able to mount an immune response against anti-TNF-α Abs, suggesting that immune complexes are a major determinant of this immunization.
Collapse
Affiliation(s)
- Christophe Arnoult
- Université François Rabelais de Tours, CNRS, UMR 7292, F-37032 Tours, France
| | - Guillaume Brachet
- Université François Rabelais de Tours, CNRS, UMR 7292, F-37032 Tours, France.,Laboratoire d'Immunologie, Centre Hospitalier Régional Universitaire de Tours, F-37032 Tours, France
| | | | - Nicolas Azzopardi
- Université François Rabelais de Tours, CNRS, UMR 7292, F-37032 Tours, France
| | - Christophe Passot
- Université François Rabelais de Tours, CNRS, UMR 7292, F-37032 Tours, France.,Laboratoire de Pharmacologie-Toxicologie, Centre Hospitalier Régional Universitaire de Tours, F-37032 Tours, France; and
| | - Celine Desvignes
- Laboratoire de Pharmacologie-Toxicologie, Centre Hospitalier Régional Universitaire de Tours, F-37032 Tours, France; and
| | - Gilles Paintaud
- Université François Rabelais de Tours, CNRS, UMR 7292, F-37032 Tours, France.,Laboratoire de Pharmacologie-Toxicologie, Centre Hospitalier Régional Universitaire de Tours, F-37032 Tours, France; and
| | - Nathalie Heuzé-Vourc'h
- Université François Rabelais de Tours, Centre d'Etude des Pathologies Respiratoires, INSERM, UMR 1100, F-37032 Tours, France
| | - Hervé Watier
- Université François Rabelais de Tours, CNRS, UMR 7292, F-37032 Tours, France.,Laboratoire d'Immunologie, Centre Hospitalier Régional Universitaire de Tours, F-37032 Tours, France
| | - Valérie Gouilleux-Gruart
- Université François Rabelais de Tours, CNRS, UMR 7292, F-37032 Tours, France; .,Laboratoire d'Immunologie, Centre Hospitalier Régional Universitaire de Tours, F-37032 Tours, France
| |
Collapse
|
3
|
Lehmann CHK, Baranska A, Heidkamp GF, Heger L, Neubert K, Lühr JJ, Hoffmann A, Reimer KC, Brückner C, Beck S, Seeling M, Kießling M, Soulat D, Krug AB, Ravetch JV, Leusen JHW, Nimmerjahn F, Dudziak D. DC subset-specific induction of T cell responses upon antigen uptake via Fcγ receptors in vivo. J Exp Med 2017; 214:1509-1528. [PMID: 28389502 PMCID: PMC5413326 DOI: 10.1084/jem.20160951] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 01/19/2017] [Accepted: 02/17/2017] [Indexed: 12/20/2022] Open
Abstract
Lehmann et al. targeted antigens to Fcγ receptors expressed on various antigen-presenting cells. Induced CD4+ and CD8+ T cell responses were solely dependent on CD11b+ and CD8+ DC subsets, respectively, but independent of receptor intrinsic ITAM or ITIM signaling domains. Dendritic cells (DCs) are efficient antigen-presenting cells equipped with various cell surface receptors for the direct or indirect recognition of pathogenic microorganisms. Interestingly, not much is known about the specific expression pattern and function of the individual activating and inhibitory Fcγ receptors (FcγRs) on splenic DC subsets in vivo and how they contribute to the initiation of T cell responses. By targeting antigens to select activating and the inhibitory FcγR in vivo, we show that antigen uptake under steady-state conditions results in a short-term expansion of antigen-specific T cells, whereas under inflammatory conditions especially, the activating FcγRIV is able to induce superior CD4+ and CD8+ T cell responses. Of note, this effect was independent of FcγR intrinsic activating signaling pathways. Moreover, despite the expression of FcγRIV on both conventional splenic DC subsets, the induction of CD8+ T cell responses was largely dependent on CD11c+CD8+ DCs, whereas CD11c+CD8− DCs were critical for priming CD4+ T cell responses.
Collapse
Affiliation(s)
- Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anna Baranska
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany.,Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, Institut National de la Santé et de la Recherche Médicale-Centre National de la Recherche Scientifique, 13288 Marseille-Luminy, France
| | - Gordon F Heidkamp
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Kirsten Neubert
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jennifer J Lühr
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Alana Hoffmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Katharina C Reimer
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Christin Brückner
- Department of Biology, Chair of Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Simone Beck
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Michaela Seeling
- Department of Biology, Chair of Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Melissa Kießling
- Department of Biology, Chair of Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Didier Soulat
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anne B Krug
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, 82152 Planegg-Martinsried, Germany
| | - Jeffrey V Ravetch
- Leonard Wagner Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10065
| | - Jeanette H W Leusen
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, 3584 Utrecht, Netherlands
| | - Falk Nimmerjahn
- Department of Biology, Chair of Genetics, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany .,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital of Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany .,Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
4
|
Abstract
Mouse and human FcRs have been a major focus of attention not only of the scientific community, through the cloning and characterization of novel receptors, and of the medical community, through the identification of polymorphisms and linkage to disease but also of the pharmaceutical community, through the identification of FcRs as targets for therapy or engineering of Fc domains for the generation of enhanced therapeutic antibodies. The availability of knockout mouse lines for every single mouse FcR, of multiple or cell-specific--'à la carte'--FcR knockouts and the increasing generation of hFcR transgenics enable powerful in vivo approaches for the study of mouse and human FcR biology. This review will present the landscape of the current FcR family, their effector functions and the in vivo models at hand to study them. These in vivo models were recently instrumental in re-defining the properties and effector functions of FcRs that had been overlooked or discarded from previous analyses. A particular focus will be made on the (mis)concepts on the role of high-affinity IgG receptors in vivo and on results from antibody engineering to enhance or abrogate antibody effector functions mediated by FcRs.
Collapse
Affiliation(s)
- Pierre Bruhns
- Unité des Anticorps en Thérapie et Pathologie, Département d'Immunologie, Institut Pasteur, Paris, France.,INSERM, U760, Paris, France
| | - Friederike Jönsson
- Unité des Anticorps en Thérapie et Pathologie, Département d'Immunologie, Institut Pasteur, Paris, France.,INSERM, U760, Paris, France
| |
Collapse
|
5
|
Wines BD, Vanderven HA, Esparon SE, Kristensen AB, Kent SJ, Hogarth PM. Dimeric FcγR Ectodomains as Probes of the Fc Receptor Function of Anti-Influenza Virus IgG. THE JOURNAL OF IMMUNOLOGY 2016; 197:1507-16. [PMID: 27385782 DOI: 10.4049/jimmunol.1502551] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 06/03/2016] [Indexed: 02/04/2023]
Abstract
Ab-dependent cellular cytotoxicity, phagocytosis, and Ag presentation are key mechanisms of action of Abs arising in vaccine or naturally acquired immunity, as well of therapeutic mAbs. Cells expressing the low-affinity FcγRs (FcγRII or CD32 and FcγRIII or CD16) are activated for these functions when receptors are aggregated following the binding of IgG-opsonized targets. Despite the diversity of the Fc receptor proteins, IgG ligands, and potential responding cell types, the induction of all FcγR-mediated responses by opsonized targets requires the presentation of multiple Fc regions in close proximity to each other. We demonstrated that such "near-neighbor" Fc regions can be detected using defined recombinant soluble (rs) dimeric low-affinity ectodomains (rsFcγR) that have an absolute binding requirement for the simultaneous engagement of two IgG Fc regions. Like cell surface-expressed FcγRs, the binding of dimeric rsFcγR ectodomains to Ab immune complexes was affected by Ab subclass, presentation, opsonization density, Fc fucosylation, or mutation. The activation of an NK cell line and primary NK cells by human IgG-opsonized influenza A hemagglutinin correlated with dimeric rsFcγRIIIa binding activity but not with Ab titer. Furthermore, the dimeric rsFcγR binding assay sensitively detected greater Fc receptor activity to pandemic H1N1 hemagglutinin after the swine influenza pandemic of 2009 in pooled human polyclonal IgG. Thus these dimeric rsFcγR ectodomains are validated, defined probes that should prove valuable in measuring the immune-activating capacity of IgG Abs elicited by infection or vaccination or experimentally derived IgG and its variants.
Collapse
Affiliation(s)
- Bruce D Wines
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia; Department of Immunology, Monash University Central Clinical School, Melbourne, Victoria 3004, Australia; Department of Pathology, The University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Hillary A Vanderven
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Sandra E Esparon
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia
| | - Anne B Kristensen
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3052, Australia; Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Melbourne, Parkville, Victoria 3052, Australia; and Melbourne Sexual Health Centre, Infectious Diseases Department, Alfred Health, Monash University Central Clinical School, Melbourne, Victoria 3004, Australia
| | - P Mark Hogarth
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria 3004, Australia; Department of Immunology, Monash University Central Clinical School, Melbourne, Victoria 3004, Australia; Department of Pathology, The University of Melbourne, Melbourne, Victoria 3052, Australia;
| |
Collapse
|
6
|
Saito T, Suenaga S, Fujii M, Kushida Y, Kawauchi Y, Suzuki K, Touma M, Hosono M. Induction of autoimmune gastritis by neonatal thymectomy requires autoantibodies and is prevented by anti-FcγR antibodies. Cell Immunol 2016; 300:1-8. [PMID: 26748859 DOI: 10.1016/j.cellimm.2015.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 09/22/2015] [Accepted: 10/21/2015] [Indexed: 11/25/2022]
Abstract
The autoantibodies (auto-Abs) that are a hallmark of neonatally thymectomized (NTx) mice with autoimmune gastritis (AIG) have been poorly explored. We investigated their immune significance using B cell-deficient (B(-)) mice and found that B(-) mice are totally resistant to AIG but become susceptible to AIG after receiving bone marrow cells from B(+) mice. This susceptibility is most likely caused by the production of auto-Abs by B cells because B(-) pups also became susceptible to AIG when nourished by an AIG dam producing auto-Abs of the IgG class during the suckling period. NTx B(-) mice receiving purified IgG auto-Abs at this developmental stage similarly developed AIG. Auto-Abs probably act on antigen handling for antigen presentation because the treatment of NTx B(+) mice with anti-FcγR Abs prevented the development of AIG. Auto-Abs are indispensable for AIG development but are not sufficient because auto-Ab treatment did not increase AIG incidence in NTx B(+) mice above the baseline.
Collapse
Affiliation(s)
- Tsubasa Saito
- Laboratory of Immunobiology, Department of Life Sciences, Graduate School of Science and Technology, Niigata University, 8050 Ikarashi 2-no-cho, Nishi-ku, Niigata 950-2181, Japan
| | - Satoru Suenaga
- Laboratory of Immunobiology, Department of Life Sciences, Graduate School of Science and Technology, Niigata University, 8050 Ikarashi 2-no-cho, Nishi-ku, Niigata 950-2181, Japan
| | - Masato Fujii
- Laboratory of Immunobiology, Department of Life Sciences, Graduate School of Science and Technology, Niigata University, 8050 Ikarashi 2-no-cho, Nishi-ku, Niigata 950-2181, Japan
| | - Yoshihiro Kushida
- Laboratory of Immunobiology, Department of Life Sciences, Graduate School of Science and Technology, Niigata University, 8050 Ikarashi 2-no-cho, Nishi-ku, Niigata 950-2181, Japan
| | - Yusuke Kawauchi
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 757 Ichibancho, Asahimachidori, Chuo-ku, Niigata 951-8510, Japan
| | - Kenji Suzuki
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Niigata University, 757 Ichibancho, Asahimachidori, Chuo-ku, Niigata 951-8510, Japan
| | - Maki Touma
- Department of Biology, Faculty of Science, Niigata University, 8050 Ikarashi 2-no-cho, Nishi-ku, Niigata 950-2181, Japan.
| | - Masamichi Hosono
- Laboratory of Immunobiology, Department of Life Sciences, Graduate School of Science and Technology, Niigata University, 8050 Ikarashi 2-no-cho, Nishi-ku, Niigata 950-2181, Japan.
| |
Collapse
|
7
|
Pyzik M, Rath T, Lencer WI, Baker K, Blumberg RS. FcRn: The Architect Behind the Immune and Nonimmune Functions of IgG and Albumin. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 194:4595-4603. [PMID: 25934922 PMCID: PMC4451002 DOI: 10.4049/jimmunol.1403014] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The neonatal FcR (FcRn) belongs to the extensive and functionally divergent family of MHC molecules. Contrary to classical MHC family members, FcRn possesses little diversity and is unable to present Ags. Instead, through its capacity to bind IgG and albumin with high affinity at low pH, it regulates the serum half-lives of both of these proteins. In addition, FcRn plays an important role in immunity at mucosal and systemic sites through its ability to affect the lifespan of IgG, as well as its participation in innate and adaptive immune responses. Although the details of its biology are still emerging, the ability of FcRn to rescue albumin and IgG from early degradation represents an attractive approach to alter the plasma half-life of pharmaceuticals. We review some of the most novel aspects of FcRn biology, immune as well as nonimmune, and provide some examples of FcRn-based therapies.
Collapse
Affiliation(s)
- Michal Pyzik
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA 02115; Department of Medicine, Harvard Medical School, Boston, MA 02115
| | - Timo Rath
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA 02115; Department of Medicine, Harvard Medical School, Boston, MA 02115; Division of Gastroenterology, Department of Medicine, Erlangen University Hospital, Friedrich Alexander University Erlangen-Nueremberg, 91054 Erlangen, Germany
| | - Wayne I Lencer
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA 02115; Harvard Digestive Diseases Center, Boston, MA 02115; and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Kristi Baker
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA 02115; Department of Medicine, Harvard Medical School, Boston, MA 02115
| | - Richard S Blumberg
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA 02115; Department of Medicine, Harvard Medical School, Boston, MA 02115; Harvard Digestive Diseases Center, Boston, MA 02115; and
| |
Collapse
|
8
|
Baker K, Rath T, Pyzik M, Blumberg RS. The Role of FcRn in Antigen Presentation. Front Immunol 2014; 5:408. [PMID: 25221553 PMCID: PMC4145246 DOI: 10.3389/fimmu.2014.00408] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 08/12/2014] [Indexed: 01/06/2023] Open
Abstract
Immunoglobulins are unique molecules capable of simultaneously recognizing a diverse array of antigens and themselves being recognized by a broad array of receptors. The abundance specifically of the IgG subclass and the variety of signaling receptors to which it binds render this an important immunomodulatory molecule. In addition to the classical Fcγ receptors that bind IgG at the cell surface, the neonatal Fc receptor (FcRn) is a lifelong resident of the endolysosomal system of most hematopoietic cells where it determines the intracellular fate of both IgG and IgG-containing immune complexes (IgG IC). Cross-linking of FcRn by multivalent IgG IC within antigen presenting cells such as dendritic cells initiates specific mechanisms that result in trafficking of the antigen-bearing IgG IC into compartments from which the antigen can successfully be processed into peptide epitopes compatible with loading onto both major histocompatibility complex class I and II molecules. In turn, this enables the synchronous activation of both CD4(+) and CD8(+) T cell responses against the cognate antigen, thereby bridging the gap between the humoral and cellular branches of the adaptive immune response. Critically, FcRn-driven T cell priming is efficient at very low doses of antigen due to the exquisite sensitivity of the IgG-mediated antigen delivery system through which it operates. FcRn-mediated antigen presentation has important consequences in tissue compartments replete with IgG and serves not only to determine homeostatic immune activation at a variety of sites but also to induce inflammatory responses upon exposure to antigens perceived as foreign. Therapeutically targeting the pathway by which FcRn enables T cell activation in response to IgG IC is thus a highly attractive prospect not only for the treatment of diseases that are driven by immune complexes but also for manipulating local immune responses against defined antigens such as those present during infections and cancer.
Collapse
Affiliation(s)
- Kristi Baker
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Timo Rath
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, Erlangen University Hospital, Friedrich Alexander University Erlangen-Nueremberg, Erlangen, Germany
| | - Michal Pyzik
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard S. Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Digestive Diseases Center, Boston, MA, USA
| |
Collapse
|
9
|
Ziętara N, Łyszkiewicz M, Krueger A, Weiss S. B-cell modulation of dendritic-cell function: signals from the far side. Eur J Immunol 2014; 44:23-32. [PMID: 24307285 DOI: 10.1002/eji.201344007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 11/26/2013] [Accepted: 11/29/2013] [Indexed: 11/05/2022]
Abstract
An appropriate immune response against a specific pathogen requires finely orchestrated interactions between the various cell populations within the immune system. At the same time, immunological tolerance to self must be maintained. DCs play an essential role in achieving these dual requisites. They coordinate adaptive immunity by integrating signals directly emanating from both infectious agents and cells of the immune system. Many such signals, especially those from innate cells and T cells, have been extensively characterized. In contrast, little is known about how B cells modulate function of DCs. B cells produce a variety of cytokines, including IL-10 and IL-6, which are known to influence DC function. In addition, Igs constitute the major secretory products of terminally differentiated B cells (plasma cells). DCs express various types of receptors for binding Ig, such as Fc receptors and C-type lectin receptors. In accordance, Igs can regulate DC function depending on the receptors engaged. Here, we review the emerging immunomodulatory role of cytokines and Ig secreted by B cells. We discuss the evidence for how these B-cell-derived factors may shape the adaptive immune response by directly acting on DCs.
Collapse
|
10
|
Baker K, Rath T, Lencer WI, Fiebiger E, Blumberg RS. Cross-presentation of IgG-containing immune complexes. Cell Mol Life Sci 2013; 70:1319-1334. [PMID: 22847331 PMCID: PMC3609906 DOI: 10.1007/s00018-012-1100-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 07/09/2012] [Accepted: 07/17/2012] [Indexed: 12/23/2022]
Abstract
IgG is a molecule that functionally combines facets of both innate and adaptive immunity and therefore bridges both arms of the immune system. On the one hand, IgG is created by adaptive immune cells, but can be generated by B cells independently of T cell help. On the other hand, once secreted, IgG can rapidly deliver antigens into intracellular processing pathways, which enable efficient priming of T cell responses towards epitopes from the cognate antigen initially bound by the IgG. While this process has long been known to participate in CD4(+) T cell activation, IgG-mediated delivery of exogenous antigens into a major histocompatibility complex (MHC) class I processing pathway has received less attention. The coordinated engagement of IgG with IgG receptors expressed on the cell-surface (FcγR) and within the endolysosomal system (FcRn) is a highly potent means to deliver antigen into processing pathways that promote cross-presentation of MHC class I and presentation of MHC class II-restricted epitopes within the same dendritic cell. This review focuses on the mechanisms by which IgG-containing immune complexes mediate such cross-presentation and the implications that this understanding has for manipulation of immune-mediated diseases that depend upon or are due to the activities of CD8(+) T cells.
Collapse
Affiliation(s)
- Kristi Baker
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115 USA
| | - Timo Rath
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115 USA
| | - Wayne I. Lencer
- Division of Gastroenterology and Nutrition, Children’s Hospital Boston, Harvard Medical School, Boston, MA 02115 USA
- Harvard Digestive Diseases Center, Boston, MA 02115 USA
| | - Edda Fiebiger
- Division of Gastroenterology and Nutrition, Children’s Hospital Boston, Harvard Medical School, Boston, MA 02115 USA
- Harvard Digestive Diseases Center, Boston, MA 02115 USA
| | - Richard S. Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115 USA
- Harvard Digestive Diseases Center, Boston, MA 02115 USA
| |
Collapse
|
11
|
Abstract
B cells have been implicated both with pathogenic as well as protective capabilities in induction and regulation of autoimmune diseases. Rheumatoid arthritis (RA) is an autoimmune disease that occurs more often in women than men. A significant role of B cells as antibody producing and antigen-presenting cells has been demonstrated in RA. Predisposition to RA is associated with the presence of certain HLA class II alleles that share sequences with DRB1*0401. To determine the role of HLA genes and B cells in vivo, we have generated transgenic mice carrying HLA genes, DRB1*0401 and DQ8, known to be associated with susceptibility to RA. Humanized mice can be induced to develop arthritis that mimics human disease in clinical, histopathological and sex bias. Effect of hormones on immune cells and their function has been described in humans and mice and has been suggested to be the major reason for female bias of autoimmune diseases. An immune response to an antigen requires presentation by HLA molecules thus suggesting a critical role of MHC in combination with sex hormones in susceptibility to develop rheumatoid arthritis. Based on our observations, we hypothesize that modulation of B cells by estrogen, presentation of modified antigens by DR4 and production of antigen-specific B cell modulating cytokines leads to autoreactivity in females. These data suggest that considering patient's sex may be crucial in selecting the optimal treatment strategy. Humanized mice expressing RA susceptible and resistant haplotype provide a means to investigate mechanism sex-bias of arthritis and future strategies for therapy.
Collapse
Affiliation(s)
- David Luckey
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | | | | |
Collapse
|
12
|
Zaharatos GJ, Yu J, Pace C, Song Y, Vasan S, Ho DD, Huang Y. HIV-1 and influenza antigens synthetically linked to IgG2a Fc elicit superior humoral responses compared to unmodified antigens in mice. Vaccine 2011; 30:42-50. [PMID: 22064264 DOI: 10.1016/j.vaccine.2011.10.056] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 10/21/2011] [Accepted: 10/23/2011] [Indexed: 01/19/2023]
Abstract
Using murine IgG subclass molecules (IgG1 or IgG2a) synthetically fused to HIV-1 or influenza test antigens, we explored the potential for IgG Fc scaffolds to augment immunogenicity. Each antigen (Ag) was grafted onto a hinge-Fc scaffold containing all critical residues necessary for interaction with effector cells, thus retaining effector functions of the native IgG subclass. We hypothesized that the differential affinity of FcγRs for specific IgG subclasses would influence the magnitude of immune responses elicited by immunization with an Ag-IgG Fc fusion vaccine. We demonstrate here that the antigen-specific humoral response elicited by Ag-IgG2a fusion vaccines is at least tenfold greater than that elicited by native antigen, that this response is superior to that elicited by Ag-IgG1, and that the augmented antigen-specific humoral response elicited is Fcγ receptor-dependent.
Collapse
Affiliation(s)
- Gerasimos J Zaharatos
- Aaron Diamond AIDS Research Center, The Rockefeller University, 455 First Avenue, 7th Floor, New York, NY 10016, USA.
| | | | | | | | | | | | | |
Collapse
|
13
|
Liu X, Lu L, Yang Z, Palaniyandi S, Zeng R, Gao LY, Mosser DM, Roopenian DC, Zhu X. The neonatal FcR-mediated presentation of immune-complexed antigen is associated with endosomal and phagosomal pH and antigen stability in macrophages and dendritic cells. THE JOURNAL OF IMMUNOLOGY 2011; 186:4674-86. [PMID: 21402891 DOI: 10.4049/jimmunol.1003584] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The FcγRs found on macrophages (Ms) and dendritic cells (DCs) efficiently facilitate the presentation or cross-presentation of immune-complexed Ags to T cells. We found that the MHC class I-related neonatal FcR for IgG (FcRn) in both Ms and DCs failed to have a strong effect on the cross-presentation of immune complex (IC) OVA Ag to CD8(+) T cells. Interestingly, endosomal FcRn enhanced the presentation of the monomeric OVA-IC to CD4(+) T cells robustly, whereas FcRn in phagosomes exerted distinctive effects on Ag presentation between Ms and DCs. The presentation of phagocytosed OVA-ICs to CD4(+) T cells was considerably enhanced on wild-type versus FcRn-deficient Ms, but was not affected in FcRn-deficient DCs. This functional discrepancy was associated with the dependence of IgG-FcRn binding in an acidic pH. Following phagocytosis, the phagosomal pH dropped rapidly to <6.5 in Ms but remained in the neutral range in DCs. This disparity in pH determined the rate of degradation of phagocytosed ICs. Thus, our findings reveal that FcRn expression has a different effect on Ag processing and presentation of ICs to CD4(+) T cells in the endosomal versus phagosomal compartments of Ms versus DCs.
Collapse
Affiliation(s)
- Xindong Liu
- Laboratory of Immunology, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Léonetti M, Gadzinski A, Moine G. Cell surface heparan sulfate proteoglycans influence MHC class II-restricted antigen presentation. THE JOURNAL OF IMMUNOLOGY 2010; 185:3847-56. [PMID: 20826758 DOI: 10.4049/jimmunol.0902724] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Heparan sulfate proteoglycans (HSPGs) are glycoproteins ubiquitously distributed on the cell surface and in the extracellular matrix. Their heparan sulfate moieties often represent alternative attachment points for extracellular proteins that target specific receptors. Thus, HSPGs modulate ligand-receptor encounters and participate in numerous biological processes. In this study, we examined whether HSPGs can also influence MHC class II-restricted Ag presentation. We selected a heparan sulfate ligand derived from the HIV-1 Tat protein and coupled it to a model protein Ag. We showed that coupling of the Tat fragment makes the Ag capable of binding cells, including APCs, and increases its ability to stimulate specific T cells up to 180-fold. The boosting effect depends on Ag processing; it vanished in the presence of an excess of heparin or free Tat fragment, indicating that HSPGs can behave as receptors involved in MHC class II processing and presentation. Furthermore, with FcγRII-bearing APCs, immune complexes containing the coupled Ag stimulated T cells up to 700-fold more efficiently than Ag-containing immune complexes. This effect vanished in the presence of heparin and is not found with FcγRII(-) APCs, indicating that HSPGs can also behave as coreceptors during FcγRIIR-mediated Ag presentation. These results indicate that ubiquitous receptors, such as HSPGs, can influence MHC class II-restricted Ag presentation and suggest that proteins will be supported more efficiently by the immune system if they have the inherent capacity to bind heparan sulfate.
Collapse
Affiliation(s)
- Michel Léonetti
- Institut de Biologie et Technologies de Saclay, Service de Pharmacologie et d' Immunoanalyse, and Laboratoire d'Ingénierie des Anticorps pour la Santé, Commissariat à l'Energie Atomique et aux Energies Alternatives, Gif-sur-Yvette, France.
| | | | | |
Collapse
|
15
|
Flores M, Desai DD, Downie M, Liang B, Reilly MP, McKenzie SE, Clynes R. Dominant expression of the inhibitory FcgammaRIIB prevents antigen presentation by murine plasmacytoid dendritic cells. THE JOURNAL OF IMMUNOLOGY 2009; 183:7129-39. [PMID: 19917701 DOI: 10.4049/jimmunol.0901169] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) are key regulators of the innate immune response, yet their direct role as APCs in the adaptive immune response is unclear. We found that unlike conventional DCs, immune complex (IC) exposed murine pDCs neither up-regulated costimulatory molecules nor activated Ag-specific CD4(+) and CD8(+) T cells. The inability of murine pDCs to promote T cell activation was due to inefficient proteolytic processing of internalized ICs. This defect in the IC processing capacity of pDCs results from a lack of activating FcgammaR expression (FcgammaRI, III, IV) and the dominant expression of the inhibitory receptor FcgammaRIIB. Consistent with this idea, transgenic expression of the activating human FcgammaRIIA gene, not present in the mouse genome, recapitulated the human situation and rescued IC antigenic presentation capacity by murine pDCs. The selective expression of FcgammaRIIB by murine pDCs was not strain dependent and was maintained even following stimulation with TLR ligands and inflammatory cytokines. The unexpected difference between the mouse and human in the expression of activating/inhibitory FcgammaRs has implications for the role of pDCs in Ab-modulated autoimmunity and anti-viral immunity.
Collapse
Affiliation(s)
- Marcella Flores
- Department of Medicine and Microbiology, Columbia-Presbyterian Medical Center, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | |
Collapse
|
16
|
Mi W, Wanjie S, Lo ST, Gan Z, Pickl-Herk B, Ober RJ, Ward ES. Targeting the neonatal fc receptor for antigen delivery using engineered fc fragments. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:7550-61. [PMID: 19017944 PMCID: PMC2738423 DOI: 10.4049/jimmunol.181.11.7550] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The development of approaches for Ag delivery to the appropriate subcellular compartments of APCs and the optimization of Ag persistence are both of central relevance for the induction of protective immunity or tolerance. The expression of the neonatal Fc receptor, FcRn, in APCs and its localization to the endosomal system suggest that it might serve as a target for Ag delivery using engineered Fc fragment-epitope fusions. The impact of FcRn binding characteristics of an Fc fragment on in vivo persistence allows this property to also be modulated. We have therefore generated recombinant Fc (mouse IgG1-derived) fusions containing the N-terminal epitope of myelin basic protein that is associated with experimental autoimmune encephalomyelitis in H-2(u) mice. The Fc fragments have distinct binding properties for FcRn that result in differences in intracellular trafficking and in vivo half-lives, allowing the impact of these characteristics on CD4(+) T cell responses to be evaluated. To dissect the relative roles of FcRn and the "classical" FcgammaRs in Ag delivery, analogous aglycosylated Fc-MBP fusions have been generated. We show that engineered Fc fragments with increased affinities for FcRn at pH 6.0-7.4 are more effective in delivering Ag to FcRn-expressing APCs in vitro relative to their lower affinity counterparts. However, higher affinity of the FcRn-Fc interaction at near neutral pH results in decreased in vivo persistence. The trade-off between improved FcRn targeting efficiency and lower half-life becomes apparent during analyses of T cell proliferative responses in mice, particularly when Fc-MBP fusions with both FcRn and FcgammaR binding activity are used.
Collapse
Affiliation(s)
- Wentao Mi
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9093
| | - Sylvia Wanjie
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9093
| | - Su-Tang Lo
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9093
| | - Zhuo Gan
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9093
| | - Beatrix Pickl-Herk
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9093
| | - Raimund J. Ober
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9093
- Department of Electrical Engineering, University of Texas at Dallas, Richardson, TX 75080
| | - E. Sally Ward
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9093
| |
Collapse
|
17
|
Desai DD, Harbers SO, Flores M, Colonna L, Downie MP, Bergtold A, Jung S, Clynes R. Fc gamma receptor IIB on dendritic cells enforces peripheral tolerance by inhibiting effector T cell responses. THE JOURNAL OF IMMUNOLOGY 2007; 178:6217-26. [PMID: 17475849 DOI: 10.4049/jimmunol.178.10.6217] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The uptake of immune complexes by FcRs on APCs augments humoral and cellular responses to exogenous Ag. In this study, CD11c+ dendritic cells are shown to be responsible in vivo for immune complex-triggered priming of T cells. We examine the consequence of Ab-mediated uptake of self Ag by dendritic cells in the rat insulin promoter-membrane OVA model and identify a role for the inhibitory FcgammaRIIB in the maintenance of peripheral CD8 T cell tolerance. Effector differentiation of diabetogenic OT-I CD8+ T cells is enhanced in rat insulin promoter-membrane OVA mice lacking FcgammaRIIB, resulting in a high incidence of diabetes. FcgammaRIIB-mediated inhibition of CD8 T cell priming results from suppression of both DC activation and cross-presentation through activating FcgammaRs. Further FcgammaRIIB on DCs inhibited the induction of OVA-specific Th1 effectors, limiting Th1-type differentiation and memory T cell accumulation. In these MHC II-restricted responses, the presence of FcgammaRIIB only modestly affected initial CD4 T cell proliferative responses, suggesting that FcgammaRIIB limited effector cell differentiation primarily by inhibiting DC activation. Thus, FcgammaRIIB can contribute to peripheral tolerance maintenance by inhibiting DC activation alone or by also limiting processing of exogenously acquired Ag.
Collapse
Affiliation(s)
- Dharmesh D Desai
- Department of Medicine and Microbiology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Taneja V, Krco CJ, Behrens MD, Luthra HS, Griffiths MM, David CS. B cells are important as antigen presenting cells for induction of MHC-restricted arthritis in transgenic mice. Mol Immunol 2007; 44:2988-96. [PMID: 17303243 PMCID: PMC1995074 DOI: 10.1016/j.molimm.2006.12.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2006] [Accepted: 12/22/2006] [Indexed: 10/23/2022]
Abstract
Rheumatoid arthritis and its animal model, collagen-induced arthritis, are known as a T and B cell dependent disease. To analyze the role of B cells in arthritis, we generated B cell deficient (microMT) mice carrying HLA-DQ8 as transgene, Abetao.DQ8.micromt mice. HLA-DQ8 transgenic mice (Abetao.DQ8) are susceptible to collagen induced arthritis, an animal model for inflammatory arthritis. Deletion of IgM gene led to the absence of B cells while T cells were comparable to Abetao.DQ8 mice. Arthritis and autoantibodies was completely abrogated in B cell deficient DQ8 mice. T cell response and proinflammatory cytokine production in response to type II collagen and its derived peptides in vitro was significantly decreased despite an increased number of Mac-1 positive cells in DQ8.micromt mice compared to DQ8 mice suggesting B cells could be important for antigen presentation as well. In vitro substitution of B cells from wild type mice restored the response in DQ8.micromt mice. B cells could also present CII-derived peptides to antigen-specific DQ8-restricted hybridomas reinforcing the role of B cells in presentation of antigens to T cells. The data suggest that B cells can be involved in pathogenesis of arthritis by producing autoantibodies and antigen presentation.
Collapse
Affiliation(s)
- Veena Taneja
- Department of Immunology, Mayo Clinic, 200 Ist Street SW, Rochester, MN 55905, USA.
| | | | | | | | | | | |
Collapse
|
19
|
Liu Y, Gao X, Masuda E, Redecha PB, Blank MC, Pricop L. Regulated Expression of FcγR in Human Dendritic Cells Controls Cross-Presentation of Antigen-Antibody Complexes. THE JOURNAL OF IMMUNOLOGY 2006; 177:8440-7. [PMID: 17142741 DOI: 10.4049/jimmunol.177.12.8440] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Receptors for IgG (FcgammaR) expressed in dendritic cells (DCs) influence the initiation of Ab-mediated immunity. Dynamic variations in FcgammaR expression allow DCs to adjust their capacity to capture Ab-opsonized Ag. The current paradigm predicts a progressive decline in FcgammaR-mediated phagocytic function upon DC maturation. Surprisingly, we find that expression of the phagocytic receptor FcgammaRIIa is preserved in immature and mature DCs at comparable levels with macrophages. Moreover, phagocytosis of antigenic peptides directed to FcgammaRIIa on DCs leads to dramatic increases in Ag cross-presentation and T cell activation. In immature DCs, high expression of inhibitory FcgammaRIIb correlates with decreased uptake and cross-presentation of Ab-Ag complexes. In contrast, engagement of FcgammaRIIb is not associated with changes in cross-presentation in mature DCs. We provide evidence that FcgammaRIIb expression is patently reduced in mature DCs, an effect that is modulated by treatment with cytokines. The regulated expression of activating and inhibitory FcgammaRs in DCs emerges as a critical checkpoint in the process of Ag uptake and cross-presentation.
Collapse
Affiliation(s)
- Yi Liu
- Research Division, Hospital for Special Surgery, Weill Graduate School of Medical Sciences, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
20
|
|
21
|
Bergtold A, Desai DD, Gavhane A, Clynes R. Cell surface recycling of internalized antigen permits dendritic cell priming of B cells. Immunity 2005; 23:503-14. [PMID: 16286018 DOI: 10.1016/j.immuni.2005.09.013] [Citation(s) in RCA: 294] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2005] [Revised: 08/29/2005] [Accepted: 09/22/2005] [Indexed: 12/30/2022]
Abstract
Dendritic cells process internalized antigens to present degradative products on MHC for TCR recognition. Because antigen-exposed DCs also induce humoral immunity, DCs must also retain antigen in its native state for the engagement of BCR on B cells. Here, we demonstrate that antigen endocytosed by the inhibitory Fc receptor, FcgammaRIIB, accesses a non-degradative intracellular vesicular compartment that recycles to the cell surface, enabling interaction of native antigen with BCR on B cells. Immunization with IgG-opsonized, T independent antigens leads to enhanced humoral responses in a FcgammaRIIB and complement dependent manner. IC-loaded DCs trafficking to the splenic marginal zone can prime a T independent response in an FcgammaRIIB-dependent manner. Thus dendritic cells are equipped with both non-degradative and degradative antigen uptake pathways to facilitate antigen presentation to both B and T cells.
Collapse
Affiliation(s)
- Amy Bergtold
- Integrated Program in Cellular, Molecular, and Biophysical Studies, Columbia University College of Physicians and Surgeons, New York, New York 10032, USA
| | | | | | | |
Collapse
|
22
|
Jarnjak-Jankovic S, Pettersen RD, Saebøe-Larssen S, Wesenberg F, Olafsen MRK, Gaudernack G. Preclinical evaluation of autologous dendritic cells transfected with mRNA or loaded with apoptotic cells for immunotherapy of high-risk neuroblastoma. Cancer Gene Ther 2005; 12:699-707. [PMID: 15846368 DOI: 10.1038/sj.cgt.7700820] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Children with high-risk neuroblastoma (NB) have a poor clinical outcome. The purpose of the present study was to evaluate different strategies for immunotherapy of high-risk NB based on vaccination with antigen-loaded dendritic cells (DCs). DCs are professional antigen-presenting cells with the ability to induce antitumor T-cell responses. We have compared DCs either loaded with apoptotic tumor cells or transfected with mRNA from the NB cell line HTB11 SK-N-SH, for their capacity to induce T-cell responses in vitro. Monocyte-derived DCs from healthy donors were loaded with tumor-derived antigens in the form of apoptotic cells or mRNA, matured and used to prime autologous T cells in vitro. After 1 week, T-cell responses against antigen-loaded DCs were measured by ELISPOT assay. DCs loaded with apoptotic NB cells or transfected with NB-cell mRNA were both able to efficiently activate autologous T cells. Both T cells of the CD8+ and CD4+ subset were activated. T cells activated by NB mRNA transfected DCs extensively crossreacted with DCs loaded with apoptotic NB cells and vice versa. The results indicate that loading of DCs with apoptotic NB cells or transfection with tumor mRNA represent promising strategies for development of individualized cancer vaccines/cancer gene therapy in treatment of NB.
Collapse
|
23
|
Baiu DC, Barger B, Sandor M, Fabry Z, Hart MN. Autoantibodies to vascular smooth muscle are pathogenic for vasculitis. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:1851-60. [PMID: 15920169 PMCID: PMC1602413 DOI: 10.1016/s0002-9440(10)62494-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We have previously shown that microvascular smooth muscle activates CD4+ T lymphocytes in sterile co-culture, presents antigen, and produces inflammatory cytokines. Adoptive transfer of lymphocytes co-cultured with syngeneic smooth muscle cells to healthy recipient mice results in vasculitic lesions predominantly in postcapillary venules. The present study assessed the pathogenic role of immunoglobulin and B cells in a murine model of vasculitis. Here, we show that transferred B cells, including plasmablast cells, accumulated, persisted, and proliferated in lung and secondary lymphoid organs of recipient mice. The induction of vasculitis was accompanied by production of IgM and IgG2a autoantibodies specific for vascular smooth muscle intracellular antigens. Circulating immunoglobulin had a pathogenic role in this vasculitis model, because the disease could be induced by transfer of serum from vasculitic mice to untreated animals but not by transfer of serum depleted of anti-smooth muscle autoantibodies. Additionally, the pathogenic mechanisms triggered by the transfer of vasculitogenic serum were dependent on T lymphocytes because both wild-type and B cell-deficient mice developed the disease after serum transfer, whereas RAG2-deficient mice did not. Thus, immunoglobulin and cell-mediated pathways work in concert to produce vasculitis in this model.
Collapse
Affiliation(s)
- Dana Carina Baiu
- Department of Pathology and Laboratory Medicine, 6152 MSC, University of Wisconsin, 1300 University Ave., Madison, WI 53706, USA
| | | | | | | | | |
Collapse
|
24
|
Kastbom A, Ahmadi A, Söderkvist P, Skogh T. The 158V polymorphism of Fc gamma receptor type IIIA in early rheumatoid arthritis: increased susceptibility and severity in male patients (the Swedish TIRA**TIRA is a Swedish acronym for ‘early invention in rheumatoid arthritis’ and is a multicentre cooperation between rheumatology units in southeastern Sweden. project). Rheumatology (Oxford) 2005; 44:1294-8. [PMID: 15998631 DOI: 10.1093/rheumatology/kei010] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVES To evaluate the influence of Fcgamma receptor IIIA (FcgammaRIIIA) 158V/F polymorphism on susceptibility and disease severity in early rheumatoid arthritis (RA). METHODS In 181 Swedish patients (128 women, 53 men) with RA of recent onset, disease and disability variables such as erythrocyte sedimentation rate, 28-joint disease activity score (DAS28) and health assessment questionnaire (HAQ) scores were monitored regularly during 3 yr. Three hundred and sixty-two controls were recruited from the same geographical area as the patients. FcgammaRIIIA genotyping was performed using denaturing high-performance liquid chromatography. RESULTS In all RA patients, FcgammaRIIIA-158VV was significantly over-represented compared with controls [odds ratio (OR) 1.9, 95% confidence interval (CI) 1.01-3.5, P<0.05]. After stratifying for sex, the difference remained in the male population (OR 3.2, 95%CI 1.03-11, P<0.05) but disappeared among women (OR 1.4, 95%CI 0.7-3.1, P=0.4). In addition, 158VV patients were more likely to exhibit early joint erosions (OR 6.1, 95%CI 1.4-28, P<0.01). At baseline, patients with different FcgammaRIIIA genotypes did not differ with respect to measures of disease activity or functional ability. Thereafter, in male patients with at least one V allele the mean DAS28 and HAQ scores were higher compared with 158FF. In contrast, female patients with at least one 158V allele displayed lower mean DAS28 and HAQ scores compared with those with 158FF. CONCLUSIONS In a male population, the FcgammaRIIIA-158VV genotype is associated with an increased risk of developing RA, and the 158V allele with more severe disease in early RA.
Collapse
Affiliation(s)
- A Kastbom
- Division of Rheumatology/AIR, Linköping University Hospital, SE-58185 Linköping, Sweden.
| | | | | | | |
Collapse
|
25
|
O'Neill SK, Shlomchik MJ, Glant TT, Cao Y, Doodes PD, Finnegan A. Antigen-specific B cells are required as APCs and autoantibody-producing cells for induction of severe autoimmune arthritis. THE JOURNAL OF IMMUNOLOGY 2005; 174:3781-8. [PMID: 15749919 DOI: 10.4049/jimmunol.174.6.3781] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
B cells play an important role in rheumatoid arthritis, but whether they are required as autoantibody-producing cells as well as APCs has not been determined. We assessed B cell autoantibody and APC functions in a murine model of autoimmune arthritis, proteoglycan (PG)-induced arthritis, using both B cell-deficient mice and Ig-deficient mice (mIgM) mice that express an H chain transgene encoding for membrane-bound, but not secreted, IgM. The IgH transgene, when paired with endogenous lambda L chain, recognizes the hapten 4-hydroxy-3-nitro-phenyl acetyl and is expressed on 1-4% of B cells. B cell-deficient and mIgM mice do not develop arthritis after immunization with PG. In adoptive transfer of PG-induced arthritis into SCID mice, T cells from mIgM mice immunized with PG were unable to transfer disease even when B cells from PG-immunized wild-type mice were provided, suggesting that the T cells were not adequately primed and that Ag-specific B cells may be required. In fact, when PG was directly targeted to the B cell Ig receptor through a conjugate of 4-hydroxy-3-nitrophenyl acetyl-PG, T cells in mIgM mice were activated and competent to transfer arthritis. Such T cells caused mild arthritis in the absence of autoantibody, demonstrating a direct pathogenic role for T cells activated by Ag-specific B cells. Transfer of arthritic serum alone induced only mild and transient arthritis. However, both autoreactive T cells and autoantibody are required to cause severe arthritis, indicating that both B cell-mediated effector pathways contribute synergistically to autoimmune disease.
Collapse
Affiliation(s)
- Shannon K O'Neill
- Department of Immunology/Microbiology, Rush University medical Center, Cohn Research Building, Chicago, IL 60612, USA
| | | | | | | | | | | |
Collapse
|
26
|
Rivera J, Zaragoza O, Casadevall A. Antibody-mediated protection against Cryptococcus neoformans pulmonary infection is dependent on B cells. Infect Immun 2005; 73:1141-50. [PMID: 15664957 PMCID: PMC546959 DOI: 10.1128/iai.73.2.1141-1150.2005] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The pathogenesis of pulmonary Cryptococcus neoformans infection and the efficacy of passive immunoglobulin G1 (IgG1) administration were investigated in B-cell-deficient and C57BL/6J mice. C57BL/6J mice lived longer than B-cell-deficient mice after both intratracheal and intravenous infections. Administration of IgG1 prior to infection prolonged the survival of C57BL/6J mice but had no effect on the survival or numbers of CFU in the lungs of B-cell-deficient mice. C. neoformans infection in B-cell-deficient mice resulted in significantly higher levels of gamma interferon (IFN-gamma), monocyte chemoattractant protein-1 (MCP-1), and macrophage inflammatory protein-1alpha (MIP-1alpha) than in C57BL/6J mice. IgG1 administration reduced IFN-gamma and MCP-1 levels in C57BL/6J mice but not in B-cell-deficient mice. In addition, compared to its effect in C57BL/6J mice, C. neoformans infection in FcRgammaIII-deficient, athymic, and SCID mice significantly increased IFN-gamma and MCP-1 levels. IgG1 administration was associated with reduced IFN-gamma levels in C57BL/6J mice but not in FcRgammaIII-deficient, athymic, and SCID mice. These observations suggest that IgG1-mediated protection in this system is a consequence of alterations in the inflammatory response that translate into less damage to the host without directly reducing the fungal burden. For hosts with impaired immunities, the ineffectiveness of passive antibody (Ab) may reflect an inability to down-regulate inflammation and avoid self-damage. The results indicate an important role for B cells in host defense against C. neoformans infection and demonstrate a surprising dependence of Ab-mediated protection on B cells in this system.
Collapse
Affiliation(s)
- Johanna Rivera
- Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | | | |
Collapse
|
27
|
Getahun A, Heyman B. IgG- and IgE-mediated antigen presentation on MHC class II. Immunol Lett 2004; 92:33-8. [PMID: 15081524 DOI: 10.1016/j.imlet.2003.09.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2003] [Accepted: 09/25/2003] [Indexed: 11/20/2022]
Abstract
IgG- and IgE-antibodies have the ability to enhance the production of antibodies directed against the antigen they are specific for. It has been suggested that the mechanism behind IgG- and IgE-mediated feedback enhancement is the ability of these isotypes to induce a more potent antigen-specific T helper cell response, increasing the chances that antigen-specific B cells receive the T cell help they require to become antibody-producing cells. With emphasis on the murine system, we will here focus on the ability of IgG and IgE to capture antigen and facilitate presentation of antigenic peptides to T helper cells. Whether this mechanism underlies feedback enhancement of antibody responses to these antigens will be discussed.
Collapse
Affiliation(s)
- Andrew Getahun
- Department of Genetics and Pathology, Uppsala University, Uppsala SE-751 85, Sweden.
| | | |
Collapse
|
28
|
Drescher B, Witte T, Schmidt RE. Glycosylation of FcgammaRIII in N163 as mechanism of regulating receptor affinity. Immunology 2003; 110:335-40. [PMID: 14632661 PMCID: PMC1783064 DOI: 10.1046/j.1365-2567.2003.01743.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human FcgammaRIII (CD16) is a low-affinity receptor for immunoglobulin G (IgG). There are two different isoforms of this protein: CD16a (transmembranous, expressed on natural killer cells and on macrophages) and CD16b (glycosylphosphatidylinositol-linked, expressed on neutrophilic granulocytes in two allelic forms NA1 and NA2). Both forms of the protein have a variable glycosylation pattern. The NA1 allele of CD16B has four asparagine (N)-linked glycosylation sites. One of them (N163) is localized in the ligand-binding site of domain II. This site is shared by the NA2 allele and CD16A. To examine the functional role of the glycosylation we mutated the four glycosylation sites of the NA1 allele (N39, N75, N163, N170) into glutamine (Q). HEK293 cells were stably transfected with the single mutants and wild-type CD16 as control. We determined binding of human IgG to transfected cells using immunofluorescence studies with anti-human IgG antibody. Monomeric IgG bound to N163Q transfectants with higher affinity than to other transfectants, showing that glycosylation in N163 influences the affinity of CD16 to its ligand. In addition, preincubation of WT-CD16-transfected cells with Tunicamycin (an inhibitor of N-glycosylation) resulted in an increased binding of monomeric IgG whereas N163Q-CD16-transfected cells remained unaffected. Therefore, glycosylation in N163 is a mechanism of regulating affinity of FcgammaRIII to its ligand IgG.
Collapse
Affiliation(s)
- Bettina Drescher
- Department of Clinical Immunology, Hannover Medical School, Carl-Neuberg-Str 1, D-30625 Hannover, Germany
| | | | | |
Collapse
|
29
|
Tan Sardjono C, Mottram PL, Hogarth PM. The role of FcgammaRIIa as an inflammatory mediator in rheumatoid arthritis and systemic lupus erythematosus. Immunol Cell Biol 2003; 81:374-81. [PMID: 12969325 DOI: 10.1046/j.1440-1711.2003.01182.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Despite their essential role in host protection, immunoglobulins are also involved in autoimmune processes where antibodies recognize the host's own tissue, triggering inflammatory responses that result in extensive tissue damage. A complex interaction of genetic predisposition, together with environment factors, is thought to trigger immune dysfunction. Although recent studies have dissected the essential role of Fc receptors in autoimmune antibody mediated processes, the uniquely human FcgammaRIIa has not been studied in detail. This Fc receptor is of particular interest, as it is the most abundantly expressed Fc receptor in humans and is implicated in immune complex disease. Investigation of its role has been hampered to date due to lack of suitable animal models. This review examines the evidence for the direct role of this receptor in diseases such as systemic lupus erythematosus and rheumatoid arthritis.
Collapse
Affiliation(s)
- Caroline Tan Sardjono
- Austin Research Institute, Kronheimer Building, Austin and Repatriation Medical Centre, Melbourne, Australia.
| | | | | |
Collapse
|
30
|
Yada A, Ebihara S, Matsumura K, Endo S, Maeda T, Nakamura A, Akiyama K, Aiba S, Takai T. Accelerated antigen presentation and elicitation of humoral response in vivo by FcγRIIB- and FcγRI/III-mediated immune complex uptake. Cell Immunol 2003; 225:21-32. [PMID: 14643301 DOI: 10.1016/j.cellimm.2003.09.008] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
It is well established that activating-type Fc receptors for IgG (FcgammaR), such as FcgammaRI and FcgammaRIII, are essential for inducing inflammatory responses, whereas a unique inhibitory FcgammaR, FcgammaRIIB, inhibits intracellular signaling upon ligation of IgG-immune complexes, and can suppress inflammation and autoimmunity. Although antigen presentation is a crucial step for evoking inflammatory responses, the contribution of FcgammaRIIB to antigen presentation is controversial as to whether it regulates antigen-presenting cells (APC), particularly dendritic cells (DC), positively or negatively. In the present report, we show that the antigen targeting to both activating-type FcgammaRs, FcgammaRI/III, and inhibitory FcgammaRIIB on bone marrow-derived DC and macrophages and primary epidermal Langerhans' cells augmented T cell proliferation in vitro and elicited humoral responses upon adoptive transfer of the antigen-pulsed DC. The DC lacking FcgammaRIIB showed a reduction in IC-uptake ability and a decreased T-cell stimulation, and induced less efficient IgG production than those of DC from wild-type mice. On the other hand, the DC lacking FcR common gamma subunit, which only expresses FcgammaRIIB, showed significant up-regulations of IC-uptake, T-cell proliferation, and IgG production compared to those of FcgammaR null DC, demonstrating a positive regulation of FcgammaRIIB for the efficient antigen presentation of IgG-complexed antigens. These results support the therapeutic benefits of antigen-targeting to FcgammaR on APC in the various inflammatory disorders.
Collapse
Affiliation(s)
- Ayumi Yada
- Department of Experimental Immunology and the CREST Program of Japan Science and Technology Corporation, Institute of Development, Aging and Cancer, Tohoku University, 980-8575, Sendai, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
den Haan JMM, Bevan MJ. Constitutive versus activation-dependent cross-presentation of immune complexes by CD8(+) and CD8(-) dendritic cells in vivo. J Exp Med 2002; 196:817-27. [PMID: 12235214 PMCID: PMC2194052 DOI: 10.1084/jem.20020295] [Citation(s) in RCA: 265] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Murine splenic dendritic cells (DCs) can be divided into two subsets based on CD8alpha expression, but the specific role of each subset in stimulation of T cells is largely unknown. An important function of DCs is the ability to take up exogenous antigens and cross-present them in the context of major histocompatibility complex (MHC) class I molecules to CD8(+) T cells. We previously demonstrated that, when cell-associated ovalbumin (OVA) is injected into mice, only the CD8(+) DC subset cross-presents OVA in the context of MHC class I. In contrast to this selectivity with cell-associated antigen, we show here that both DC subsets isolated from mice injected with OVA/anti-OVA immune complexes (OVA-IC) cross-present OVA to CD8(+) T cells. The use of immunoglobulin G Fc receptor (Fc(gamma)R) common gamma-chain-deficient mice revealed that the cross-presentation by CD8(-) DCs depended on the expression of gamma-chain-containing activating FcgammaRs, whereas cross-presentation by CD8(+) DCs was not reduced in gamma-chain-deficient mice. These results suggest that although CD8(+) DCs constitutively cross-present exogenous antigens in the context of MHC class I molecules, CD8(-) DCs only do so after activation, such as via ligation of Fc(gamma)Rs. Cross-presentation of immune complexes may play an important role in autoimmune diseases and the therapeutic effect of antitumor antibodies.
Collapse
Affiliation(s)
- Joke M M den Haan
- Department of Immunology, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195-7370, USA
| | | |
Collapse
|
32
|
Rafiq K, Bergtold A, Clynes R. Immune complex–mediated antigen presentation induces tumor immunity. J Clin Invest 2002. [DOI: 10.1172/jci0215640] [Citation(s) in RCA: 182] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
33
|
Rafiq K, Bergtold A, Clynes R. Immune complex-mediated antigen presentation induces tumor immunity. J Clin Invest 2002; 110:71-9. [PMID: 12093890 PMCID: PMC151032 DOI: 10.1172/jci15640] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Antigen uptake receptors on dendritic cells (DCs) provide efficient entry for the initiation of antigen-specific adaptive immunity. Here we show that targeting of antigen to Fc receptors on DCs accomplishes combined activation of Th1 CD4 and CD8 effector responses in vivo, namely delayed-type hypersensitivity and tumor immunity. Tumor immunity specific for ovalbumin-expressing tumors was provided by immunization with wild-type but not FcgammaRgamma(-/-) DCs loaded with ovalbumin-containing immune complexes. Tumor protection was eliminated when immune complex-loaded DCs lacked beta(2) microglobulin, TAP, or MHC class II, demonstrating that Fc receptor-targeted antigenic uptake led to both MHC class I- and class II-restricted responses, which together are required for effector tumor immunity. Thus the cross-presentation pathway accessed by antigens acquired endocytically through Fc receptors links humoral and cellular immunity. These data suggest that administration of antitumor antibodies may enhance tumor-specific T cell responses in vivo and provide the rationale for Fc receptor targeting in vaccine development.
Collapse
MESH Headings
- Animals
- Antibodies, Neoplasm/biosynthesis
- Antigen Presentation
- Antigen-Antibody Complex/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/immunology
- Dendritic Cells/immunology
- Histocompatibility Antigens Class I/metabolism
- Histocompatibility Antigens Class II/metabolism
- Immunization, Passive
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Mice
- Mice, Congenic
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Ovalbumin/immunology
- Receptors, IgE/genetics
- Receptors, IgE/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Khadija Rafiq
- Department of Medicine and Microbiology, Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | | | | |
Collapse
|
34
|
Dhodapkar KM, Krasovsky J, Williamson B, Dhodapkar MV. Antitumor monoclonal antibodies enhance cross-presentation ofcCellular antigens and the generation of myeloma-specific killer T cells by dendritic cells. J Exp Med 2002; 195:125-33. [PMID: 11781371 PMCID: PMC2196013 DOI: 10.1084/jem.20011097] [Citation(s) in RCA: 284] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The mechanism of antitumor effect of monoclonal antibodies (mAbs) is not fully understood. Here we show that coating myeloma cells with anti-syndecan-1 antibody promotes cross-presentation of cellular antigens by dendritic cells (DCs) to autologous T cells from healthy donors. The tumor cells treated with anti-syndecan-1 or isotype-matched control antibody were fed to HLA-mismatched monocyte-derived immature DCs. Tumor cell-loaded mature DCs induced a strong CD8(+) T cell response that was specific for the cancer-testis (C-T) antigens expressed in the tumor. The CD8(+) T cells killed peptide-pulsed targets, as well as myeloma tumor cells. Importantly, mAbs-coated tumor-loaded DCs were consistently superior to DCs loaded with peptides or dying cells for eliciting tumor-specific killer T cells. This enhanced cross-presentation was not due to enhanced tumor cell uptake or to DC maturation. When mixtures of NY-Eso-1-positive and -negative myeloma cells were captured by DCs, the anti-syndecan-1 antibody had to be on the NY-Eso-1-positive cells to elicit NY-Eso-1-specific response. Cross-presentation was inhibited by pretreatment of DCs with Fc gamma receptor blocking antibodies. Targeting of mAb-coated tumors to DCs may contribute to the efficacy of tumor-reactive mAb and offers a new strategy for immunotherapy.
Collapse
Affiliation(s)
- Kavita M Dhodapkar
- Laboratory of Tumor Immunology and Immunotherapy, The Rockefeller University, New York, NY 10021, USA
| | | | | | | |
Collapse
|
35
|
Abdul-Majid KB, Stefferl A, Bourquin C, Lassmann H, Linington C, Olsson T, Kleinau S, Harris RA. Fc receptors are critical for autoimmune inflammatory damage to the central nervous system in experimental autoimmune encephalomyelitis. Scand J Immunol 2002; 55:70-81. [PMID: 11841694 DOI: 10.1046/j.1365-3083.2002.01024.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Multiple sclerosis (MS) is simulated by various forms of experimental autoimmune encephalomyelitis, in which T cells, antibodies, cytokines and complementary factors interact with the central nervous system (CNS) myelin proteins and lead to inflammatory damage. We investigated the role of Fc receptors (FcRs), which link the cellular and humoral branches of the immune system, in myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE), using two different FcRgamma knockout DBA/1 mice. The first knockout were the FcRgamma chain-deficient mice, which lack FcgammaRI, FcgammaRIII and Fc(epsilon)RI, while the second knockout mice lack only FcgammaRII. The lack of FcgammaRII enhanced the disease susceptibility with associated increased CNS demyelination. While FcRgamma+/+ DBA/1 mice also developed pronounced CNS infiltration and myelin destruction, FcRgamma-/- littermates were protected despite initial peripheral autoimmune responses to MOG. In vitro analyses revealed equivalent potentials of fluid phase phagocytosis of myelin and MOG in bone-marrow macrophages derived from both FcRgamma+/+ and FcRgamma-/- mice, while MOG-immunoglobulin (Ig)G immune complexes were only internalized by FcRgamma+/+ macrophages. This was associated with cellular activation in FcRgamma+/+ but not FcRgamma-/- macrophages, as assessed by the activation of intracellular mitogen activated protein (MAP)-kinase signalling elements. We propose that protection from EAE in FcRgamma-deficient mice is due to the inefficient antigen processing/presentation of myelin proteins during the induction of secondary immune responses locally in the CNS, which leads to demyelination. This demonstrates the importance of FcR in the promotion of autoimmune inflammation of the CNS and highlights the therapeutic possibility of treatment of MS with FcR-directed modalities.
Collapse
MESH Headings
- Animals
- Antigen Presentation
- Autoantibodies/blood
- Autoimmunity
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Humans
- In Vitro Techniques
- Macrophage Activation
- Macrophages/immunology
- Mice
- Mice, Inbred DBA
- Mice, Knockout
- Multiple Sclerosis/etiology
- Multiple Sclerosis/immunology
- Multiple Sclerosis/therapy
- Myelin Proteins
- Myelin-Associated Glycoprotein/immunology
- Myelin-Oligodendrocyte Glycoprotein
- Phagocytosis
- Receptors, IgG/genetics
- Receptors, IgG/metabolism
- Spinal Cord/immunology
- Spinal Cord/pathology
Collapse
Affiliation(s)
- K-B Abdul-Majid
- Neuroimmunology Unit, L8:04 Center for Molecular Medicine, Karolinska Hospital, SE-17176 Stockholm, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Crowe JE. Influence of maternal antibodies on neonatal immunization against respiratory viruses. Clin Infect Dis 2001; 33:1720-7. [PMID: 11595986 DOI: 10.1086/322971] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2001] [Revised: 05/17/2001] [Indexed: 11/03/2022] Open
Abstract
Vaccines that successfully prevent severe infant respiratory virus diseases should induce protection at a very young age because of the low age of patients who are hospitalized owing to these viruses. Candidate respiratory virus vaccines are being tested in infants who are naïve to infection but seropositive to the viral agents because they possess maternal IgG antibodies (Abs). Transplacental maternal Abs may be partially protective against disease caused by respiratory virus infections. Carefully conducted studies have shown that these Abs can also profoundly suppress or enhance infant immune responses to immunization. The mechanisms underlying regulation of immune responses to viruses by maternal Abs are under investigation. This article explores the current knowledge regarding the effect of maternal Abs on respiratory virus and measles virus immunization, and it reviews the current approaches to overcoming Ab-mediated immunosuppression.
Collapse
Affiliation(s)
- J E Crowe
- Departments of Pediatrics and Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
37
|
Märten A, Ziske C, Schöttker B, Renoth S, Weineck S, Buttgereit P, Schakowski F, von Rücker A, Sauerbruch T, Schmidt-Wolf IG. Interactions between dendritic cells and cytokine-induced killer cells lead to an activation of both populations. J Immunother 2001; 24:502-10. [PMID: 11759073 DOI: 10.1097/00002371-200111000-00007] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dendritic cells (DCs) are major antigen-presenting cells. They are capable of capturing and processing tumor antigens, expressing lymphocyte costimulatory molecules, and secreting cytokines to initiate immune responses. Here, the authors tested the effect of cytokine-induced killer (CIK) cells, a population that includes CD3+CD56+ cells (natural killer T cells), with regard to their capacity to immunomodulate DCs. Cytokine-induced killer cells were cocultured with autologous DCs generated from peripheral blood mononuclear cells. Expression of markers typical for both populations was measured using flow cytometry, and secretion of interleukin (IL)-12 was determined using enzyme-linked immunosorbent assays. Cytotoxicity assays were performed to investigate the role of IL-12 and the importance of cell-cell interactions. Considering this, receptors for IL-12 and CD40 were blocked and cocultures were performed with cell culture inserts. Coculture of CIK cells led to a significant increase of DC-specific, costimulatory, and antigen-presenting molecules in DC cultures. In addition, coculture resulted in a dramatically increase of IL-12 secretion by DCs and to a significant increase in cytotoxic activity of CIK cells toward carcinoma cells. Blockage of IL-12 uptake decreased the cytolytic activity of CIK cells. Cytokine secretion was shown to be important for activation of CIK cells, and also cellular interactions between DCs and effector cells caused a higher cytolytic capacity. Interactions between DCs and CIK cells caused changes in the surface molecule expression of both populations, led to an increase of IL-12 secretion, and rendered an improved cytotoxic activity. The natural killer T cell subpopulation seems to be responsible for this effect. Therefore, coculture of DCs with CIK cells may have a major impact on immunotherapeutic protocols for patients with cancer.
Collapse
Affiliation(s)
- A Märten
- Medizinische Klinik und Poliklinik I, Allgemeine Innere Medizin, Rheinische Friedrich-Wilhelms-Universität, Bonn, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Fogg MH, Parsons KR, Thomas LH, Taylor G. Identification of CD4+ T cell epitopes on the fusion (F) and attachment (G) proteins of bovine respiratory syncytial virus (BRSV). Vaccine 2001; 19:3226-40. [PMID: 11312019 DOI: 10.1016/s0264-410x(01)00030-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
To gain insight into the antigenic structure of the F and G proteins of BRSV, we have mapped CD4+ T cell epitopes on these proteins using synthetic peptides and lymphocytes from vaccinated, naturally infected or experimentally infected calves, in proliferation assays. Bovine CD4+ T cells recognised epitopes that were distributed predominantly within the F1 subunit of the F protein, some of which were adjacent to previously identified B cell epitopes. Bovine CD4+ T cell epitopes within the G protein were mainly located within the cytoplasmic tail. Several immunodominant bovine T cell epitopes within the F protein, that were recognised by calves with different haplotypes, are also recognised by human T cells. Thus, cattle and humans appear to recognise similar T cell epitopes on the F protein. Studies using antibodies to bovine MHC class II and BoLA DR-transfected CHO cells as antigen-presenting cells indicated that immunodominant regions of the F and G proteins contained both DR- and DQ-restricted epitopes. The finding that there was little recognition of the extracellular domain of the G protein by T cells has important implications for vaccine design based on the soluble form of this protein.
Collapse
Affiliation(s)
- M H Fogg
- Institute for Animal Health, Compton, Berkshire, RG20 7NN, Newbury, UK
| | | | | | | |
Collapse
|
39
|
Runyon K, Lee K, Zuberek K, Collins M, Leonard JP, Dunussi-Joannopoulos K. The combination of chemotherapy and systemic immunotherapy with soluble B7-immunoglobulin G leads to cure of murine leukemia and lymphoma and demonstration of tumor-specific memory responses. Blood 2001; 97:2420-6. [PMID: 11290606 DOI: 10.1182/blood.v97.8.2420] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Major mechanisms underlying poor immune responses to autologous tumor-associated antigens are overwhelming tumor kinetics and the absence of effective T-cell costimulation by antigen-presenting cells. To address these issues, leukemia and lymphoma mice were treated with the combination of chemotherapy and systemic immunotherapy with recombinant soluble murine B7-immunoglobulin G (IgG) molecules. In this report, 3 murine models were used, a radiation-induced SJL acute myeloid leukemia, a transplantable spontaneous SJL lymphoma, and the C57BL/6 EL-4 thymic lymphoma. Various treatment modalities were evaluated: single treatments with either B7-IgG or chemotherapy as well as combination therapies. The results demonstrate the following: (1) in all tumor models, the combination of chemotherapy and soluble B7-IgGs is more potent than either therapy alone, leading to cure of tumor-bearing animals; (2) the therapeutic responses are T-cell-dependent, because combined therapy is not efficacious in severe combined immunodeficient mice; (3) the rejection of tumor cells leads to the development of tumor-specific immunity, because cured mice are immune to the rejected tumor but not to a different syngeneic tumor; and (4) (51)Cr release assays show that rejection of tumor cells leads to the development of very potent tumor-specific cytotoxic T-lymphocyte activity. On the basis of these results, it is proposed that chemotherapy-mediated tumor reduction, together with consequent augmented tumor-antigen presentation to activated T cells, are primary mechanisms leading to curative responses. The safety profile of the B7-IgG fusion proteins and their synergy with chemotherapy strongly suggest that the combination regimen is a promising strategy in cancer treatment.
Collapse
MESH Headings
- Acute Disease
- Animals
- Antigens, CD/genetics
- Antigens, CD/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- B7-1 Antigen/genetics
- B7-1 Antigen/therapeutic use
- B7-2 Antigen
- Combined Modality Therapy
- Cyclophosphamide/administration & dosage
- Cytarabine/administration & dosage
- Doxorubicin/administration & dosage
- Female
- Immunoglobulin G/genetics
- Immunoglobulin G/therapeutic use
- Immunologic Memory
- Immunotherapy
- Leukemia, Myeloid/drug therapy
- Leukemia, Myeloid/immunology
- Leukemia, Myeloid/therapy
- Leukemia, Radiation-Induced/drug therapy
- Leukemia, Radiation-Induced/immunology
- Leukemia, Radiation-Induced/therapy
- Lymphoma, Non-Hodgkin/drug therapy
- Lymphoma, Non-Hodgkin/immunology
- Lymphoma, Non-Hodgkin/therapy
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/therapeutic use
- Mice
- Mice, Inbred C57BL
- Mice, SCID
- Neoplasm Transplantation
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/therapeutic use
- T-Lymphocytes/immunology
- Thymus Neoplasms/drug therapy
- Thymus Neoplasms/immunology
- Thymus Neoplasms/therapy
Collapse
Affiliation(s)
- K Runyon
- Genetics Institute, Wyeth-Ayerst Research, Cambridge, MA, USA
| | | | | | | | | | | |
Collapse
|
40
|
Affiliation(s)
- J Pieters
- Basel Institute for Immunology, Switzerland
| |
Collapse
|
41
|
Stockmeyer B, Dechant M, van Egmond M, Tutt AL, Sundarapandiyan K, Graziano RF, Repp R, Kalden JR, Gramatzki M, Glennie MJ, van de Winkel JG, Valerius T. Triggering Fc alpha-receptor I (CD89) recruits neutrophils as effector cells for CD20-directed antibody therapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:5954-61. [PMID: 11067958 DOI: 10.4049/jimmunol.165.10.5954] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD20 Abs induce clinical responses in lymphoma patients, but there are considerable differences between individual patients. In (51)Cr release assays with whole blood as effector source, RAJI cells were effectively killed by a mouse/human chimeric IgG1 construct of CD20 Ab 1F5, whereas ARH-77 proved resistant to killing by this Ab. When whole blood was fractionated into plasma, mononuclear cells, or granulocytic effector cells, RAJI cells were effectively killed in the presence of complement-containing plasma, whereas the mature B cell line ARH-77 proved complement resistant. However, with a bispecific Ab (BsAb) against the myeloid receptor for IgA (CD89; FcalphaRI) and CD20, a broad range of B cell lines were effectively killed. FcalphaRI is expressed on monocytes/macrophages, neutrophils, and eosinophils. As the numbers of these effector cells and their functional activity can be enhanced by application of G-CSF or GM-CSF, lysis via (FcalphaRI x CD20) BsAb was significantly enhanced in blood from patients during therapy with these myeloid growth factors. Interestingly, the major effector cell population for this BsAb were polymorphonuclear neutrophils, which proved ineffective in killing malignant B cells with murine, chimeric IgG1, or FcgammaRI- or FcgammaRIII-directed BsAbs against CD20. Experiments with blood from human FcalphaRI/FcgammaRI double-transgenic mice showed corresponding results, allowing the establishment of relevant syngenic animal models in these mice. In conclusion, the combination of myeloid growth factors and an (FcalphaRI x CD20) BsAb may represent a promising approach to improve effector cell recruitment for CD20-directed lymphoma therapy.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/genetics
- Antibodies, Bispecific/pharmacology
- Antibodies, Bispecific/therapeutic use
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Neoplasm/genetics
- Antibodies, Neoplasm/pharmacology
- Antibody-Dependent Cell Cytotoxicity/genetics
- Antigens, CD/biosynthesis
- Antigens, CD/blood
- Antigens, CD/physiology
- Antigens, CD20/immunology
- Antineoplastic Agents/pharmacology
- Granulocyte Colony-Stimulating Factor/pharmacology
- Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology
- Growth Inhibitors/pharmacology
- Humans
- Immunoglobulin A/metabolism
- Immunoglobulin G/genetics
- Immunoglobulin G/pharmacology
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/therapy
- Mice
- Mice, Transgenic
- Neutrophil Infiltration/immunology
- Receptors, Fc/biosynthesis
- Receptors, Fc/blood
- Receptors, Fc/physiology
- Receptors, IgG/biosynthesis
- Receptors, IgG/blood
- Receptors, IgG/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/pharmacology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- B Stockmeyer
- Division of Hematology/Oncology, Department of Medicine III, University of Erlangen-Nürnberg, Erlangen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Shen L, Lang ML, Wade WF. The ins and outs of getting in: structures and signals that enhance BCR or Fc receptor-mediated antigen presentation. IMMUNOPHARMACOLOGY 2000; 49:227-40. [PMID: 10996020 DOI: 10.1016/s0162-3109(00)00255-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Antigen-presenting cells internalize antigen by fluid-phase pinocytosis or by endocytosis via surface receptors such as the B cell receptor (BCR) and Fc receptors for IgG, IgA and IgE (FcR). While both modes of internalization lead to antigen presentation it is recognized that receptor-mediated endocytosis greatly enhances the efficiency of processing and antigen presentation. Receptors facilitate the entry of antigen into the endocytic pathway by interaction of their internalization motifs with the endocytic machinery. These motifs include tyrosine-based, dileucine and casein kinase-like motifs. However these structures appear insufficient to support processing of cryptic epitopes, leading to a limited immune response. Cryptic epitope processing appears dependent on receptor signaling which is mediated by immunoreceptor tyrosine activation motifs (ITAMs). The signaling cascade which follows receptor crosslinking promotes reorganization and acidification of the late endocytic compartment or MIIC. Signaling events downstream of Syk, in particular calcium flux and protein kinase C activation, are necessary for MIIC induction. PI(3) kinase is also involved at multiple steps in antigen presentation, including production of PIP3 and transport of cathepsins. PIP3 is crucial both as a binding substrate for proteins implicated in vesicle transport and for the recruitment of signaling molecules to the plasma membrane. Among PIP3 activated molecules, protein kinase B (PKB) has been linked to endocytic function. We observe association of activated PKB with the MIIC after signaling through antigen presentation-competent receptors, but not mutant, presentation-defective receptors.
Collapse
Affiliation(s)
- L Shen
- Department of Microbiology, Dartmouth Medical School, Lebanon, NH 03756, USA
| | | | | |
Collapse
|
43
|
Hioe CE, Jones GJ, Rees AD, Ratto-Kim S, Birx D, Münz C, Gorny MK, Tuen M, Zolla-Pazner S. Anti-CD4-binding domain antibodies complexed with HIV type 1 glycoprotein 120 inhibit CD4+ T cell-proliferative responses to glycoprotein 120. AIDS Res Hum Retroviruses 2000; 16:893-905. [PMID: 10875615 DOI: 10.1089/08892220050042837] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
HIV-specific CD4+ helper T cell responses, particularly to the envelope glycoproteins, are usually weak or absent in the majority of HIV-seropositive individuals. Since antibodies, by their capacity to alter antigen uptake and processing, are known to have modulatory effects on CD4+ T cell responses, we investigated the effect of antibodies produced by HIV-infected individuals on the CD4+ T cell response to HIV-1 gp120. Proliferative responses of gp120-specific CD4+ T cells were inhibited in the presence of either serum immunoglobulin from HIV-infected individuals or human monoclonal antibodies specific for the CD4-binding domain (CD4bd) of gp120. Human monoclonal antibodies to other gp120 epitopes did not have the same effect. The anti-CD4bd antibodies complexed with gp120 suppressed T cell lines specific for varying gp120 epitopes but did not affect T cell proliferation to non-HIV antigens. Moreover, inhibition by the anti-CD4bd/gp120 complexes was observed regardless of the types of antigen-presenting cells used to stimulate the T cells. These results indicate that the presence of anti-CD4bd antibodies complexed with gp120 can strongly suppress CD4+ helper T responses to gp120.
Collapse
Affiliation(s)
- C E Hioe
- New York Veteran Affairs Medical Center and Department of Pathology, New York University School of Medicine, New York 10010, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Foucras G, Gapin L, Coureau C, Kanellopoulos JM, Guéry JC. Interleukin 4-producing CD4 T cells arise from different precursors depending on the conditions of antigen exposure in vivo. J Exp Med 2000; 191:683-94. [PMID: 10684860 PMCID: PMC2195842 DOI: 10.1084/jem.191.4.683] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
The precursor origin of T helper (Th) cell subsets in vivo has been difficult to study and remains poorly investigated. We have previously shown that chronic administration of soluble protein antigen induces selective development of antigen-specific CD4 Th2 cells in genetically predisposed mouse strains. To analyze the origin of effector T cells in this model, we designed a competitive polymerase chain reaction-based approach to track public BV-J rearrangement expressed by CD4 T cells specific for hen egg white lysozyme (HEL) in BALB/c mice. We show that public T cell clones are predominantly associated with type 1 or 2 effector Th cells recovered after primary immunization in complete or incomplete Freund's adjuvant, respectively. Conversely, continuous administration of soluble antigen, which induces strong memory Th2 response, is associated with a dose-dependent reduction of public clone size by a mechanism resembling clonal anergy. Thus, soluble HEL-induced Th2 cells do not express the public complementarity determining region 3 motifs characteristic of immunogenic challenge in the presence of adjuvant. These results demonstrate that there are multiple pathways of induction of Th2 responses depending on the condition of antigen exposure in vivo, i.e., clonal immune deviation versus recruitment of a different pool of precursor cells.
Collapse
Affiliation(s)
- Gilles Foucras
- Institut National de la Santé et de la Recherche Médicale (INSERM) U28, Institut Fédératif de Recherche 30, Hôpital Purpan, 31059 Toulouse Cedex, France
| | - Laurent Gapin
- Unité de Biologie Moléculaire du Gène, INSERM U277, Institut Pasteur, 75724 Paris cedex, France
| | - Christiane Coureau
- Institut National de la Santé et de la Recherche Médicale (INSERM) U28, Institut Fédératif de Recherche 30, Hôpital Purpan, 31059 Toulouse Cedex, France
| | - Jean M. Kanellopoulos
- Unité de Biologie Moléculaire du Gène, INSERM U277, Institut Pasteur, 75724 Paris cedex, France
| | - Jean-Charles Guéry
- Institut National de la Santé et de la Recherche Médicale (INSERM) U28, Institut Fédératif de Recherche 30, Hôpital Purpan, 31059 Toulouse Cedex, France
| |
Collapse
|
45
|
Bakke O, Nordeng TW. Intracellular traffic to compartments for MHC class II peptide loading: signals for endosomal and polarized sorting. Immunol Rev 1999; 172:171-87. [PMID: 10631946 DOI: 10.1111/j.1600-065x.1999.tb01365.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In this review we focus on the traffic of MHC class II and endocytosed antigens to intracellular compartments where antigenic peptides are loaded. We also discuss briefly the nature of the peptide loading compartment and the sorting signals known to direct antigen receptors and MHC class II and associated molecules to this location. MHC class II molecules are expressed on a variety of polarized epithelial and endothelial cells, and polarized cells are thus potentially important for antigen presentation. Here we review some cell biological aspects of polarized sorting of MHC class II and the associated invariant chain and the signals that are involved in the sorting process to the basolateral domain. The molecules involved in sorting and loading of peptide may modulate antigen presentation, and in particular we discuss how invariant chain may change the cellular phenotype and the kinetics of the endosomal pathway.
Collapse
Affiliation(s)
- O Bakke
- Department of Biology, University of Oslo, Norway.
| | | |
Collapse
|
46
|
Abstract
Antigens internalized through specific membrane receptors are presented to helper CD4(+) T cells at antigen concentrations 10(3) to 10(4) fold lower than antigens internalized by fluid phase. B lymphocyte antigen receptors, mannose receptors and receptors for the Fc region of immunoglobulins, promote both internalization and efficient presentation at low antigen concentrations. Thus, binding to specific membrane receptors concentrate antigens on antigen presenting cells and mediates efficient uptake. Is this 'quantitative' concentration of antigens on antigen presenting cells the end of the story? Or may 'quality', i.e. selective intracellular antigen targeting, somehow influence the efficiency or specificity of MHC class I and class II-restricted antigen presentation?
Collapse
Affiliation(s)
- S Amigorena
- INSERM U520, Institut Curie, 12 rue Lhomond, Paris, 75005, France
| | | |
Collapse
|
47
|
van Bergen J, Ossendorp F, Jordens R, Mommaas AM, Drijfhout JW, Koning F. Get into the groove! Targeting antigens to MHC class II. Immunol Rev 1999; 172:87-96. [PMID: 10631939 DOI: 10.1111/j.1600-065x.1999.tb01358.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The activation of MHC class II-restricted helper T cells is paramount to adaptive immune responses. Vaccine development could therefore benefit from improved ways of targeting antigens into MHC class II molecules. In recent years, the natural pathways of MHC class II antigen presentation have been exploited to achieve this goal. First, antigenic proteins and peptides have been modified to facilitate receptor-mediated uptake by professional antigen-presenting cells. Second, DNA constructs containing specific targeting sequences have been used to direct endogenously synthesized antigens to the MHC class II compartments. Both strategies proved to be highly effective. We review these data and describe how this knowledge is currently applied to the design of vaccines that activate helper T cells in vivo.
Collapse
Affiliation(s)
- J van Bergen
- Dept of Immunohematology, Leiden University Medical Center, The Netherlands.
| | | | | | | | | | | |
Collapse
|
48
|
Abstract
Most hematopoietic cells express a wide variety of receptors for the Fc portion of immunoglobulins (FcR) belonging to the immunoreceptor family. FcRs are multichain complexes composed of ligand-binding alpha chains, which determine Ig binding, and signal tranduction subunits, bearing a conserved immunoreceptor tyrosine-based activation motif (ITAM). Besides signaling, most Fc gamma Rs also efficiently internalize antigen-antibodies complexes and thus induce efficient processing of antigens into peptides presented by major histocompatibility complex (MHC) class II molecules. Importantly, ITAMs and cytosolic effectors of cell signaling also determine Fc gamma R's endocytic transport and antigen presentation capacities.
Collapse
Affiliation(s)
- S Amigorena
- U520 INSERM, Institut Curie, Section de Recherche, Paris, France.
| | | |
Collapse
|
49
|
Dong X, Storkus WJ, Salter RD. Binding and Uptake of Agalactosyl IgG by Mannose Receptor on Macrophages and Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.10.5427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Increased levels of agalactosyl IgG (G0 IgG) are found in several autoimmune diseases, including rheumatoid arthritis, in which they are correlated with severity of the disease. To investigate whether structural alteration of IgG may lead to aberrant processing and presentation of IgG peptides as autoantigens, we have studied uptake of G0 IgG by human dendritic cells and macrophages cultured from PBMC. We found that enzymatic removal of terminal galactose residues, which exposes N-acetylglucosamine residues, increases uptake of soluble IgG mediated by mannose receptor on macrophages and dendritic cells. Efficient uptake appears to require recycling of the receptor, can be blocked by saccharides or Abs reactive with mannose receptor, and is dependent upon the state of maturation of the dendritic cells. No differences between IgG isotypes in ability to be internalized by APC were identified, suggesting that uptake would not be limited to a particular subset of Abs. These results suggest a novel pathway by which Abs or Ag-Ab complexes can be taken into dendritic cells and macrophages, and potentially generate epitopes recognized by T cells. These findings may have particular relevance for autoimmune disorders characterized by high levels of G0 IgG.
Collapse
Affiliation(s)
| | - Walter J. Storkus
- †Surgery, and
- ‡University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Russell D. Salter
- *Pathology and
- ‡University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| |
Collapse
|
50
|
Elsässer D, Stadick H, van de Winkel JG, Valerius T. GM-CSF as adjuvant for immunotherapy with bispecific antibodies. Eur J Cancer 1999; 35 Suppl 3:S25-8. [PMID: 10645218 DOI: 10.1016/s0959-8049(99)00086-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- D Elsässer
- Department of Urology, University of Erlangen-Nürnberg, Germany
| | | | | | | |
Collapse
|