1
|
Simpson AP, Oldham RJ, Cox KL, Taylor MC, James S, White AL, Bogdanov Y, Glennie MJ, Frendeus B, Cragg MS, Roghanian A. FcγRIIB (CD32B) antibodies enhance immune responses through activating FcγRs. Clin Exp Immunol 2025; 219:uxaf015. [PMID: 40089806 PMCID: PMC12046127 DOI: 10.1093/cei/uxaf015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/20/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025] Open
Abstract
Fc receptors (FcR) play a key role in coordinating responses from both the innate and adaptive immune system. The inhibitory Fc gamma receptor (FcγRIIB/CD32B; referred to as FcγRII/CD32 in mice) restrains the immune response, specifically through regulating immunoglobulin G (IgG) effector functions. FcγRII-deficient mice demonstrate elevated incidence and severity of autoimmunity and increased responses to immunization and infections. To explore the potential of FcγRIIB as a target for augmenting vaccines, we tested the ability of monoclonal antibodies (mAb) against mouse FcγRII and human FcγRIIB to enhance humoral responses in preclinical models. We used wild-type (WT), FcγR-deficient, and human FcγRIIB transgenic (Tg) mice with either a functional intracellular domain (hFcγRIIB Tg) or lacking immunoreceptor tyrosine-based inhibitory motif (ITIM) signalling capacity (NoTIM). Targeting mouse FcγRII and human FcγRIIB with antibodies significantly augmented humoral immune responses against experimental antigens and enhanced tumour clearance in vivo. Surprisingly, mAbs without a functional Fc (N297Q; referred to as Fc-null) lacked efficacy. Similarly, blocking FcγRII in mice lacking activating FcγRs failed to enhance immune responses. Conversely, blocking both signalling-competent and signalling-defective (NoTIM) FcγRIIB in Tg mice with a WT, but not Fc-null, FcγRIIB mAb equally enhanced immunity. These data indicate the redundancy of inhibitory signalling in potentiating immune responses in vivo. Collectively, our data suggest that mAb-targeting of FcγRIIB stabilizes mAb Fc and enhances immune responses via Fc-mediated crosslinking of activating FcγRs, irrespective of the inhibitory function of FcγRIIB. These findings support a strategy to boost immune responses in immunization protocols.
Collapse
Affiliation(s)
- Alexander P Simpson
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
| | - Robert J Oldham
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
| | - Kerry L Cox
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
| | - Martin C Taylor
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
| | - Sonya James
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
| | - Ann L White
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
| | - Yury Bogdanov
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
| | - Martin J Glennie
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
| | - Björn Frendeus
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
- BioInvent International AB, Sölvegatan 41, Lund, Sweden
| | - Mark S Cragg
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
- Institute for Life Sciences, University of Southampton, Highfield, Southampton, UK
| | - Ali Roghanian
- Antibody and Vaccine Group, School of Cancer Sciences, Centre for Cancer Immunology, University of Southampton Faculty of Medicine, Southampton, Hampshire, UK
- Institute for Life Sciences, University of Southampton, Highfield, Southampton, UK
| |
Collapse
|
2
|
Douglas TR, Alexander S, Chou LYT. Patterned Antigens on DNA Origami Controls the Structure and Cellular Uptake of Immune Complexes. ACS NANO 2025; 19:621-637. [PMID: 39757925 DOI: 10.1021/acsnano.4c11183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Immune complexes (ICs), formed via antibody (Ab)-antigen (Ag) binding, trigger diverse immune responses, which are critical for natural immunity and have uses for vaccines and immunotherapies. While IC-elicited immune responses depend on its structure, existing methods for IC synthesis produce heterogeneous assemblies, which limits control over their cellular interactions and pharmacokinetics. In this study, we demonstrate the use of DNA origami to create synthetic ICs with defined shape, size, and solubility by displaying Ags in prescribed spatial patterns. We find that Ag arrangement relative to the spatial tolerance of IgG Fab arms (∼13-18 nm) determines IC formation into "monomeric" versus "multimeric" regimes. When Ag spacing matches Fab arm tolerance, ICs are exclusively monomeric, while spacing mismatches favor the formation of multimeric ICs. Within each IC regime, parameters such as the number of Ags and Ab-Ag ratios, as well as DNA origami shape, further fine-tune IC size, shape, and Fc valency. These parameters influenced IC interactions with FcγR-expressing immune cells, with uptake by macrophages showing greater sensitivity to IC cross-linking while dendritic cells were more responsive to Ab valency. Our findings thus provide design principles for controlling the structure and cellular interactions of synthetic ICs and highlight DNA origami-scaffolded ICs as a programmable platform for investigating IC immunology and developing FcγR-targeted therapeutics and vaccines.
Collapse
Affiliation(s)
- Travis R Douglas
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 2E3, Canada
| | - Shana Alexander
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 2E3, Canada
| | - Leo Y T Chou
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 2E3, Canada
| |
Collapse
|
3
|
Rohrer B, Parsons N, Annamalai B, Nicholson C, Obert E, Jones BW, Dick AD. Peptide-based immunotherapy against oxidized elastin ameliorates pathology in mouse model of smoke-induced ocular injury. Exp Eye Res 2021; 212:108755. [PMID: 34487725 PMCID: PMC9753162 DOI: 10.1016/j.exer.2021.108755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Age-related macular degeneration (AMD), the leading cause of blindness in western populations, is associated with an overactive complement system, and an increase in circulating antibodies against certain epitopes, including elastin. As loss of the elastin layer of Bruch's membrane (BrM) has been reported in aging and AMD, we previously showed that immunization with elastin peptide oxidatively modified by cigarette smoke (ox-elastin), exacerbated ocular pathology in the smoke-induced ocular pathology (SIOP) model. Here we asked whether ox-elastin peptide-based immunotherapy (PIT) ameliorates damage. METHODS C57BL/6J mice were injected with ox-elastin peptide at two doses via weekly subcutaneous administration, while exposed to cigarette smoke for 6 months. FcγR-/- and uninjected C57BL/6J mice served as controls. Retinal morphology was assessed by electron microscopy, and complement activation, antibody deposition and mechanisms of immunological tolerance were assessed by Western blotting and ELISA. RESULTS Elimination of Fcγ receptors, preventing antigen/antibody-dependent cytotoxicity, protected against SIOP. Mice receiving PIT with low dose ox-elastin (LD-PIT) exhibited reduced humoral immunity, reduced complement activation and IgG/IgM deposition in the RPE/choroid, and largely a preserved BrM. While there is no direct evidence of ox-elastin pathogenicity, LD-PIT reduced IFNγ and increased IL-4 within RPE/choroid. High dose PIT was not protective. CONCLUSIONS These data further support ox-elastin role in ocular damage in part via elastin-specific antibodies, and support the corollary that PIT with ox-elastin attenuates ocular pathology. Overall, damage is associated with complement activation, antibody-dependent cell-mediated cytotoxicity, and altered cytokine signature.
Collapse
Affiliation(s)
- Bärbel Rohrer
- Departments of Ophthalmology and Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA; Departments of Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA; Departments of Ralph H. Johnson VA Medical Center, Division of Research, Charleston, SC, 29401, USA.
| | - Nathaniel Parsons
- Departments of Ophthalmology and Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA
| | - Balasubramaniam Annamalai
- Departments of Ophthalmology and Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA
| | - Crystal Nicholson
- Departments of Ophthalmology and Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA
| | - Elisabeth Obert
- Departments of Ophthalmology and Neurosciences Division of Research, Medical University of South Carolina, Charleston, SC, USA
| | - Bryan W Jones
- Department of Ophthalmology, University of Utah, Salt Lake City, UT, 84132, USA
| | - Andrew D Dick
- University of Bristol, Bristol BS8 1TD, UK and University College London-Institute of Ophthalmology and the National Institute for Health Research Biomedical Research Centre, Moorfields Eye Hospital, London, EC1V 9EL, UK.
| |
Collapse
|
4
|
Kdimati S, Mullins CS, Linnebacher M. Cancer-Cell-Derived IgG and Its Potential Role in Tumor Development. Int J Mol Sci 2021; 22:11597. [PMID: 34769026 PMCID: PMC8583861 DOI: 10.3390/ijms222111597] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/13/2021] [Accepted: 10/23/2021] [Indexed: 12/18/2022] Open
Abstract
Human immunoglobulin G (IgG) is the primary component of the human serum antibody fraction, representing about 75% of the immunoglobulins and 10-20% of the total circulating plasma proteins. Generally, IgG sequences are highly conserved, yet the four subclasses, IgG1, IgG2, IgG3, and IgG4, differ in their physiological effector functions by binding to different IgG-Fc receptors (FcγR). Thus, despite a similarity of about 90% on the amino acid level, each subclass possesses a unique manner of antigen binding and immune complex formation. Triggering FcγR-expressing cells results in a wide range of responses, including phagocytosis, antibody-dependent cell-mediated cytotoxicity, and complement activation. Textbook knowledge implies that only B lymphocytes are capable of producing antibodies, which recognize specific antigenic structures derived from pathogens and infected endogenous or tumorigenic cells. Here, we review recent discoveries, including our own observations, about misplaced IgG expression in tumor cells. Various studies described the presence of IgG in tumor cells using immunohistology and established correlations between high antibody levels and promotion of cancer cell proliferation, invasion, and poor clinical prognosis for the respective tumor patients. Furthermore, blocking tumor-cell-derived IgG inhibited tumor cells. Tumor-cell-derived IgG might impede antigen-dependent cellular cytotoxicity by binding antigens while, at the same time, lacking the capacity for complement activation. These findings recommend tumor-cell-derived IgG as a potential therapeutic target. The observed uniqueness of Ig heavy chains expressed by tumor cells, using PCR with V(D)J rearrangement specific primers, suggests that this specific part of IgG may additionally play a role as a potential tumor marker and, thus, also qualify for the neoantigen category.
Collapse
Affiliation(s)
| | | | - Michael Linnebacher
- Clinic of General Surgery, Molecular Oncology and Immunotherapy, University Medical Center Rostock, University of Rostock, 18057 Rostock, Germany; (S.K.); (C.S.M.)
| |
Collapse
|
5
|
Macri C, Morgan H, Villadangos JA, Mintern JD. Regulation of dendritic cell function by Fc-γ-receptors and the neonatal Fc receptor. Mol Immunol 2021; 139:193-201. [PMID: 34560415 DOI: 10.1016/j.molimm.2021.07.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 06/28/2021] [Accepted: 07/19/2021] [Indexed: 01/02/2023]
Abstract
Dendritic cells (DCs) express receptors to sense pathogens and/or tissue damage and to communicate with other immune cells. Among those receptors, Fc receptors (FcRs) are triggered by the Fc region of antibodies produced during adaptive immunity. In this review, the role of FcγR and neonatal Fc receptor (FcRn) in DC immunity will be discussed. Their expression in DC subsets and impact on antigen uptake and presentation, DC maturation and polarisation of T cell responses will be described. Lastly, we will discuss the importance of FcR-mediated DC function in the context of immunity during viral infection, inflammatory disease, cancer and immunotherapy.
Collapse
Affiliation(s)
- Christophe Macri
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia
| | - Huw Morgan
- ACRF Translational Research Laboratory, The Royal Melbourne Hospital, Parkville, Melbourne, Victoria, 3050, Australia; Department of Medicine, University of Melbourne, Parkville, Melbourne, Victoria, 3010, Australia
| | - Jose A Villadangos
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia; Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, 3010, Australia.
| | - Justine D Mintern
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
6
|
Wiedinger K, McCauley J, Bitsaktsis C. Isotype-specific outcomes in Fc gamma receptor targeting of PspA using fusion proteins as a vaccination strategy against Streptococcus pneumoniae infection. Vaccine 2020; 38:5634-5646. [PMID: 32646816 DOI: 10.1016/j.vaccine.2020.06.067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/22/2020] [Accepted: 06/22/2020] [Indexed: 02/08/2023]
Abstract
Streptococcus pneumoniae (Spn) remains a considerable threat to public health despite the availability of antibiotics and polysaccharide conjugate vaccines. The lack of mucosal immunity in addition to capsular polysaccharide diversity, has proved to be problematic in developing a universal vaccine against Spn. Targeting antigen to Fc receptors is an attractive way to augment both innate and adaptive immunity against mucosal pathogens, by promoting interactions with activating Fcγ receptors (FcγR) that mediate diverse immunomodulatory functions. The effect of targeting FcγR is highly influenced by the IgG subclass, which bares differential affinities for activating and inhibitory FcγR. In the current study we demonstrate targeting activating FcγR with fusion proteins consisting of PspA and IgG2a Fc enhance PspA-specific immune responses, and effectively protect against mucosal Spn challenge. Specifically, targeting PspA to FcγR polarized alveolar macrophage to the AM1 phenotype and increased conventional dendritic cell subsets in the lung in addition to augmenting Th1 cytokines and PspA-specific IgG and IgA. In contrast, fusion proteins consisting of PspA fused to the IgG1 Fc provided minimal benefit over administration of PspA alone, as a result of interaction with the inhibitory FcγRIIB. Protective efficacy of the IgG1 fusion protein was significantly enhanced in animals deficient for FcγRIIB accompanied by increased B cell maturation and proliferation levels in these animals. These studies demonstrate FcγR targeting is an effective strategy for inducing potent cellular and humoral responses via mucosal immunization with Fc fusion proteins, however, careful consideration of the Fc region utilized is required since Fc isotype subclass heavily influenced immunization induced effector functions and survival against lethal Spn challenge. Fc-engineering with specific attention to FcγRIIB engagement presents a valuable vaccine strategy for protecting against Spn infection.
Collapse
Affiliation(s)
- Kari Wiedinger
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, USA.
| | - James McCauley
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, USA.
| | | |
Collapse
|
7
|
Danzer H, Glaesner J, Baerenwaldt A, Reitinger C, Lux A, Heger L, Dudziak D, Harrer T, Gessner A, Nimmerjahn F. Human Fcγ-receptor IIb modulates pathogen-specific versus self-reactive antibody responses in lyme arthritis. eLife 2020; 9:55319. [PMID: 32613944 PMCID: PMC7438111 DOI: 10.7554/elife.55319] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022] Open
Abstract
Pathogen-specific antibody responses need to be tightly regulated to generate protective but limit self-reactive immune responses. While loss of humoral tolerance has been associated with microbial infections, the pathways involved in balancing protective versus autoreactive antibody responses in humans are incompletely understood. Studies in classical mouse model systems have provided evidence that balancing of immune responses through inhibitory receptors is an important quality control checkpoint. Genetic differences between inbred mouse models and the outbred human population and allelic receptor variants not present in mice; however, argue for caution when directly translating these findings to the human system. By studying Borrelia burgdorferi infection in humanized mice reconstituted with human hematopoietic stem cells from donors homozygous for a functional or a non-functional FcγRIIb allele, we show that the human inhibitory FcγRIIb is a critical checkpoint balancing protective and autoreactive immune responses, linking infection with induction of autoimmunity in the human immune system.
Collapse
Affiliation(s)
- Heike Danzer
- Institute of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Joachim Glaesner
- Institute of Medical Microbiology and Hygiene, University Regensburg, Regensburg, Germany
| | - Anne Baerenwaldt
- Laboratory for Cancer Immunotherapy, University Hospital Basel, Basel, Switzerland
| | - Carmen Reitinger
- Institute of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Anja Lux
- Institute of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Lukas Heger
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen, Erlangen, Germany
| | - Diana Dudziak
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen, Erlangen, Germany.,Medical Immunology Campus Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Thomas Harrer
- Medical Department 3, University Hospital Erlangen, Erlangen, Germany
| | - André Gessner
- Institute of Medical Microbiology and Hygiene, University Regensburg, Regensburg, Germany
| | - Falk Nimmerjahn
- Institute of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany.,Medical Immunology Campus Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
8
|
Morita M, Kajiye M, Sakurai C, Kubo S, Takahashi M, Kinoshita D, Hori N, Hatsuzawa K. Characterization of MORN2 stability and regulatory function in LC3-associated phagocytosis in macrophages. Biol Open 2020; 9:bio051029. [PMID: 32414768 PMCID: PMC7327995 DOI: 10.1242/bio.051029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/28/2020] [Indexed: 01/25/2023] Open
Abstract
Microtubule-associated protein A1/B1-light chain 3 (LC3)-associated phagocytosis (LAP) is a type of non-canonical autophagy that regulates phagosome maturation in macrophages. However, the role and regulatory mechanism of LAP remain largely unknown. Recently, the membrane occupation and recognition nexus repeat-containing-2 (MORN2) was identified as a key component of LAP for the efficient formation of LC3-recruiting phagosomes. To characterize MORN2 and elucidate its function in LAP, we established a MORN2-overexpressing macrophage line. At a steady state, MORN2 was partially cleaved by the ubiquitin-proteasome system. MORN2 overexpression promoted not only LC3-II production but also LAP phagosome (LAPosome) acidification during Escherichia coli uptake. Furthermore, the formation of LAPosomes containing the yeast cell wall component zymosan was enhanced in MORN2-overexpressing cells and depended on reactive oxygen species (ROS). Finally, MORN2-mediated LAP was regulated by plasma membrane-localized soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) such as SNAP-23 and syntaxin 11. Taken together, these findings demonstrate that MORN2, whose expression is downregulated via proteasomal digestion, is a limiting factor for LAP, and that membrane trafficking by SNARE proteins is involved in MORN2-mediated LAP.
Collapse
Affiliation(s)
- Maya Morita
- Division of Molecular Biology, School of Life Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan
| | - Mayu Kajiye
- Division of Molecular Biology, School of Life Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan
| | - Chiye Sakurai
- Division of Molecular Biology, School of Life Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan
| | - Shuichi Kubo
- Division of Molecular Biology, School of Life Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan
| | - Miki Takahashi
- Division of Molecular Biology, School of Life Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan
| | - Daiki Kinoshita
- Division of Molecular Biology, School of Life Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan
| | - Naohiro Hori
- Division of Molecular Biology, School of Life Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan
| | - Kiyotaka Hatsuzawa
- Division of Molecular Biology, School of Life Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan
| |
Collapse
|
9
|
Cormier M, Batty P, Tarrant J, Lillicrap D. Advances in knowledge of inhibitor formation in severe haemophilia A. Br J Haematol 2020; 189:39-53. [DOI: 10.1111/bjh.16377] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Matthew Cormier
- Department of Pathology and Molecular Medicine Richardson Laboratory Queen’s University Kingston ON Canada
| | - Paul Batty
- Department of Pathology and Molecular Medicine Richardson Laboratory Queen’s University Kingston ON Canada
| | - Julie Tarrant
- Department of Pathology and Molecular Medicine Richardson Laboratory Queen’s University Kingston ON Canada
| | - David Lillicrap
- Department of Pathology and Molecular Medicine Richardson Laboratory Queen’s University Kingston ON Canada
| |
Collapse
|
10
|
Ritprajak P, Kaewraemruaen C, Hirankarn N. Current Paradigms of Tolerogenic Dendritic Cells and Clinical Implications for Systemic Lupus Erythematosus. Cells 2019; 8:cells8101291. [PMID: 31640263 PMCID: PMC6830089 DOI: 10.3390/cells8101291] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/05/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022] Open
Abstract
Tolerogenic dendritic cells (tolDCs) are central players in the initiation and maintenance of immune tolerance and subsequent prevention of autoimmunity. Recent advances in treatment of autoimmune diseases including systemic lupus erythematosus (SLE) have focused on inducing specific tolerance to avoid long-term use of immunosuppressive drugs. Therefore, DC-targeted therapies to either suppress DC immunogenicity or to promote DC tolerogenicity are of high interest. This review describes details of the typical characteristics of in vivo and ex vivo tolDC, which will help to select a protocol that can generate tolDC with high functional quality for clinical treatment of autoimmune disease in individual patients. In addition, we discuss the recent studies uncovering metabolic pathways and their interrelation intertwined with DC tolerogenicity. This review also highlights the clinical implications of tolDC-based therapy for SLE treatment, examines the current clinical therapeutics in patients with SLE, which can generate tolDC in vivo, and further discusses on possibility and limitation on each strategy. This synthesis provides new perspectives on development of novel therapeutic approaches for SLE and other autoimmune diseases.
Collapse
Affiliation(s)
- Patcharee Ritprajak
- Research Unit in Integrative Immuno-Microbial Biochemistry and Bioresponsive Nanomaterials, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand.
- Department of Microbiology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Chamraj Kaewraemruaen
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Nattiya Hirankarn
- Center of Excellence in Immunology and Immune-Mediated Diseases, Chulalongkorn University, Bangkok 10330, Thailand.
- Immunology Unit, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
11
|
Verbeek JS, Hirose S, Nishimura H. The Complex Association of FcγRIIb With Autoimmune Susceptibility. Front Immunol 2019; 10:2061. [PMID: 31681256 PMCID: PMC6803437 DOI: 10.3389/fimmu.2019.02061] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 08/15/2019] [Indexed: 12/20/2022] Open
Abstract
FcγRIIb is the only inhibitory Fc receptor and controls many aspects of immune and inflammatory responses. The observation 19 years ago that Fc γ RIIb -/- mice generated by gene targeting in 129 derived ES cells developed severe lupus like disease when backcrossed more than 7 generations into C57BL/6 background initiated extensive research on the functional understanding of this strong autoimmune phenotype. The genomic region in the distal part of Chr1 both in human and mice in which the Fc γ R gene cluster is located shows a high level of complexity in relation to the susceptibility to SLE. Specific haplotypes of closely linked genes including the Fc γ RIIb and Slamf genes are associated with increased susceptibility to SLE both in mice and human. Using forward and reverse genetic approaches including in human GWAS and in mice congenic strains, KO mice (germline and cell type specific, on different genetic background), knockin mice, overexpressing transgenic mice combined with immunological models such as adoptive transfer of B cells from Ig transgenic mice the involved genes and the causal mutations and their associated functional alterations were analyzed. In this review the results of this 19 years extensive research are discussed with a focus on (genetically modified) mouse models.
Collapse
Affiliation(s)
- J Sjef Verbeek
- Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| | - Sachiko Hirose
- Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| | - Hiroyuki Nishimura
- Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| |
Collapse
|
12
|
Marvin J, Rhoads JP, Major AS. FcγRIIb on CD11c + cells modulates serum cholesterol and triglyceride levels and differentially affects atherosclerosis in male and female Ldlr -/- mice. Atherosclerosis 2019; 285:108-119. [PMID: 31051414 DOI: 10.1016/j.atherosclerosis.2019.04.221] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Circulating levels of oxidized lipoprotein (oxLDL) correlate with myocardial infarction risk and atherosclerosis severity. Our previous study demonstrates that oxLDL immune complexes (oxLDL-ICs) can signal through FcγRs on bone marrow-derived dendritic cells (BMDCs) and enhance their activation and inflammatory cytokine secretion. While global FcγR-/- studies have shown that activating FcγRs are proatherogenic, the role of the inhibitory FcγRIIb is unclear. We sought to determine the role of DC-specific FcγRIIb in atherosclerosis. METHODS Bone marrow chimeras were generated by rescuing lethally irradiated Ldlr-/- mice with hematopoietic cells from littermate CD11c-Cre+ or CD11c-Cre-Fcgr2bfl/fl donors. Four weeks following transplant, recipients were placed on a Western diet for eight weeks. Various tissues and organs were analyzed for differences in inflammation. RESULTS Quantitation of atherosclerosis in the proximal aorta demonstrated a 58% increase in female CD11c-Cre+Fcgr2bfl/fl recipients, but a surprising 44% decrease in male recipients. Hepatic cholesterol and triglycerides were increased in female CD11c-Cre+Fcgr2bfl/fl recipients. This was associated with an increase in CD36 and MHC Class II expression on hepatic CD11c+CD11b+ DCs in female livers. In contrast, male CD11c-Cre+Fcgr2bfl/fl recipients had decreased hepatic lipids with a corresponding decrease in CD36 and MHC Class II expression on CD11c+ cells. Interestingly, both sexes of CD11c-Cre+Fcgr2bfl/fl recipients had significant decreases in serum cholesterol and TGs with corresponding decreases in liver Fasn transcripts. CONCLUSIONS The absence of FcγRIIb expression on CD11c+ cells results in sex-dependent alteration in liver inflammation influencing atherogenesis and sex-independent modulation of serum cholesterol and TGs.
Collapse
Affiliation(s)
- Jennifer Marvin
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt Medical Center, Nashville, TN, 37232, USA
| | - Jillian P Rhoads
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt Medical Center, Nashville, TN, 37232, USA
| | - Amy S Major
- Tennessee Valley Healthcare System, U.S. Department of Veterans Affairs, Nashville, TN, 37212, USA; Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
13
|
Teige I, Mårtensson L, Frendéus BL. Targeting the Antibody Checkpoints to Enhance Cancer Immunotherapy-Focus on FcγRIIB. Front Immunol 2019; 10:481. [PMID: 30930905 PMCID: PMC6423481 DOI: 10.3389/fimmu.2019.00481] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/22/2019] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy with therapeutic antibodies has increased survival for patients with hematologic and solid cancers. Still, a significant fraction of patients fails to respond to therapy or acquire resistance. Understanding and overcoming mechanisms of resistance to antibody drugs, and in particular those common to antibody drugs as a class, is therefore highly warranted and holds promise to improve response rates, duration of response and potentially overall survival. Activating and inhibitory Fc gamma receptors (FcγR) are known to coordinately regulate therapeutic activity of tumor direct-targeting antibodies. Similar, but also divergent, roles for FcγRs in controlling efficacy of immune modulatory antibodies e.g., checkpoint inhibitors have been indicated from mouse studies, and were recently implicated in contributing to efficacy in the human clinical setting. Here we discuss evidence and mechanisms by which Fc gamma receptors–the “antibody checkpoints”–regulate antibody-induced antitumor immunity. We further discuss how targeted blockade of the sole known inhibitory antibody checkpoint FcγRIIB may help overcome resistance and boost activity of clinically validated and emerging antibodies in cancer immunotherapy.
Collapse
|
14
|
Funes SC, Manrique de Lara A, Altamirano-Lagos MJ, Mackern-Oberti JP, Escobar-Vera J, Kalergis AM. Immune checkpoints and the regulation of tolerogenicity in dendritic cells: Implications for autoimmunity and immunotherapy. Autoimmun Rev 2019; 18:359-368. [PMID: 30738957 DOI: 10.1016/j.autrev.2019.02.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The immune system is responsible for defending the host from a large variety of potential pathogens, while simultaneously avoiding immune reactivity towards self-components. Self-tolerance has to be tightly maintained throughout several central and peripheral processes; immune checkpoints are imperative for regulating the immunity/tolerance balance. Dendritic cells (DCs) are specialized cells that capture antigens, and either activate or inhibit antigen-specific T cells. Therefore, they play a key role at inducing and maintaining immune tolerance. DCs that suppress the immune response have been called tolerogenic dendritic cells (tolDCs). Given their potential as a therapy to prevent transplant rejection and autoimmune damage, several strategies are under development to generate tolDCs, in order to avoid activation and expansion of self-reactive T cells. In this article, we summarize the current knowledge relative to the main features of tolDCs, their mechanisms of action and their therapeutic use for autoimmune diseases. Based on the literature reviewed, autologous antigen-specific tolDCs might constitute a promising strategy to suppress autoreactive T cells and reduce detrimental inflammatory processes.
Collapse
Affiliation(s)
- Samanta C Funes
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Amaranta Manrique de Lara
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituto de Biotecnología, Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico.
| | - María J Altamirano-Lagos
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Juan P Mackern-Oberti
- Instituto de Medicina y Biología Experimental de Cuyo, IMBECU, CONICET, Mendoza, Argentina; Instituto de Fisiología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.
| | - Jorge Escobar-Vera
- Laboratorio de Genética, Departamento Biomédico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile.
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Endocrinología, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
15
|
Lu Q, Grotzke JE, Cresswell P. A novel probe to assess cytosolic entry of exogenous proteins. Nat Commun 2018; 9:3104. [PMID: 30082832 PMCID: PMC6079096 DOI: 10.1038/s41467-018-05556-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 07/06/2018] [Indexed: 01/08/2023] Open
Abstract
Dendritic cells use a specialized pathway called cross-presentation to activate CD8+ T cells by presenting peptides from exogenous protein antigens on major histocompatibility complex class I molecules. Considerable evidence suggests that internalized antigens cross endocytic membranes to access cytosolic proteasomes for processing. The mechanism of protein dislocation represents a major unsolved problem. Here we describe the development of a sensitive reporter substrate, an N-glycosylated variant of Renilla luciferase fused to the Fc region of human IgG1. The luciferase variant is designed to be enzymatically inactive when glycosylated, but active after the asparagine to aspartic acid conversion that occurs upon deglycosylation by the cytosolic enzyme N-glycanase-1. The generation of cytosolic luminescence depends on internalization, deglycosylation, the cytosolic AAA-ATPase VCP/p97, and the cytosolic chaperone HSP90. By incorporating a T cell epitope into the fusion protein, we demonstrate that antigen dislocation into the cytosol is the rate limiting step in cross-presentation.
Collapse
Affiliation(s)
- Qiao Lu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Jeff E Grotzke
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Peter Cresswell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
16
|
Roghanian A, Stopforth RJ, Dahal LN, Cragg MS. New revelations from an old receptor: Immunoregulatory functions of the inhibitory Fc gamma receptor, FcγRIIB (CD32B). J Leukoc Biol 2018; 103:1077-1088. [PMID: 29406570 DOI: 10.1002/jlb.2mir0917-354r] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/03/2017] [Accepted: 12/14/2017] [Indexed: 12/11/2022] Open
Abstract
The Fc gamma receptor IIB (FcγRIIB/CD32B) was generated million years ago during evolution. It is the sole inhibitory receptor for IgG, and has long been associated with the regulation of humoral immunity and innate immune homeostasis. However, new and surprising functions of FcγRIIB are emerging. In particular, FcγRIIB has been shown to perform unexpected activatory roles in both immune-signaling and monoclonal antibody (mAb) immunotherapy. Furthermore, although ITIM signaling is an integral part of FcγRIIB regulatory activity, it is now clear that inhibition/activation of immune responses can occur independently of the ITIM. In light of these new findings, we present an overview of the established and noncanonical functions of FcγRIIB and discuss how this knowledge might be exploited therapeutically.
Collapse
Affiliation(s)
- Ali Roghanian
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Richard J Stopforth
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Lekh N Dahal
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Mark S Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| |
Collapse
|
17
|
Abstract
IgG antibodies mediate a diversity of immune functions by coupling of antigen specificity through the Fab domain to signal transduction via Fc-Fc receptor interactions. Indeed, balanced IgG signaling through type I and type II Fc receptors is required for the control of proinflammatory, anti-inflammatory, and immunomodulatory processes. In this review, we discuss the mechanisms that govern IgG-Fc receptor interactions, highlighting the diversity of Fc receptor-mediated effector functions that regulate immunity and inflammation as well as determine susceptibility to infection and autoimmunity and responsiveness to antibody-based therapeutics and vaccines.
Collapse
Affiliation(s)
- Stylianos Bournazos
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York 10065;
| | - Taia T Wang
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York 10065;
| | - Rony Dahan
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York 10065;
| | - Jad Maamary
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York 10065;
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York 10065;
| |
Collapse
|
18
|
Fcγ Receptor Function and the Design of Vaccination Strategies. Immunity 2017; 47:224-233. [PMID: 28813656 DOI: 10.1016/j.immuni.2017.07.009] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/17/2017] [Indexed: 02/07/2023]
Abstract
Through specific interactions with distinct types of Fcγ receptors (FcγRs), the Fc domain of immunoglobulin G (IgG) mediates a wide spectrum of immunological functions that influence both innate and adaptive responses. Recent studies indicate that IgG Fc-FcγR interactions are dynamically regulated during an immune response through the control of the Fc-associated glycan structure and Ig subclass composition on the one hand and selective FcγR expression on immune cells on the other, which together determine the capacity of IgG to interact in a cell-type-specific manner with specific members of the FcγR family. Here, we present a framework that synthesizes the current understanding of the contribution of FcγR pathways to the induction and regulation of antibody and T cell responses. Within this context, we discuss vaccination strategies to elicit broad and potent immune responses based on the immunomodulatory properties of Fc-FcγR interactions.
Collapse
|
19
|
Abstract
The antiviral activity of antibodies reflects the bifunctional properties of these molecules. While the Fab domains mediate highly specific antigenic recognition to block virus entry, the Fc domain interacts with diverse types of Fcγ receptors (FcγRs) expressed on the surface of effector leukocytes to induce the activation of distinct immunomodulatory pathways. Fc-FcγR interactions are tightly regulated to control IgG-mediated inflammation and immunity and are largely determined by the structural heterogeneity of the IgG Fc domain, stemming from differences in the primary amino acid sequence of the various subclasses, as well as the structure and composition of the Fc-associated N-linked glycan. Engagement of specific FcγR types on effector leukocytes has diverse consequences that affect several aspects of innate and adaptive immunity. In this review, we provide an overview of the complexity of FcγR-mediated pathways, discussing their role in the in vivo protective activity of anti-HIV-1 antibodies. We focus on recent studies on broadly neutralizing anti-HIV-1 antibodies that revealed that Fc-FcγR interactions are required to achieve full therapeutic activity through clearance of IgG-opsonized virions and elimination of HIV-infected cells. Manipulation of Fc-FcγR interactions to specifically activate distinct FcγR-mediated pathways has the potential to affect downstream effector responses, influencing thereby the in vivo protective activity of anti-HIV-1 antibodies; a strategy that has already been successfully applied to other IgG-based therapeutics, substantially improving their clinical efficacy.
Collapse
Affiliation(s)
- Stylianos Bournazos
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY, USA
| | - Jeffrey V Ravetch
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY, USA
| |
Collapse
|
20
|
Abstract
A key determinant for the survival of organisms is their capacity to recognize and respond efficiently to foreign antigens. This is largely accomplished by the orchestrated activity of the innate and adaptive branches of the immune system. Antibodies are specifically generated in response to foreign antigens, facilitating thereby the specific recognition of antigens of almost infinite diversity. Receptors specific for the Fc domain of antibodies, Fc receptors, are expressed on the surface of the various myeloid leukocyte populations and mediate the binding and recognition of antibodies by innate leukocytes. By directly linking the innate and the adaptive components of immunity, Fc receptors play a central role in host defense and the maintenance of tissue homeostasis through the induction of diverse proinflammatory, anti-inflammatory, and immunomodulatory processes that are initiated upon engagement by the Fc domain. In this chapter, we discuss the mechanisms that regulate Fc domain binding to the various types of Fc receptors and provide an overview of the astonishing diversity of effector functions that are mediated through Fc-FcR interactions on myeloid cells. Lastly, we discuss the impact of FcR-mediated interactions in the context of IgG-mediated inflammation, autoimmunity, susceptibility to infection, and responsiveness to antibody-based therapeutics.
Collapse
|
21
|
Bournazos S, Ravetch JV. Diversification of IgG effector functions. Int Immunol 2017; 29:303-310. [PMID: 28472280 PMCID: PMC5890892 DOI: 10.1093/intimm/dxx025] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 04/26/2017] [Indexed: 12/16/2022] Open
Abstract
IgG is the major immunoglobulin class produced during an immune response against foreign antigens and efficiently provides protection through its bifunctional nature. While the Fab domains confer highly specific recognition of the antigen, the Fc domain mediates a wide range of effector functions that modulate several aspects of innate and adaptive immunity. Engagement of the various types of Fcγ receptors (FcγRs) by an IgG Fc domain can activate distinct immunomodulatory pathways with pleiotropic functional consequences for several leukocyte types. Fc effector functions are not limited to phagocytosis and cytotoxicity of IgG-opsonized targets but exhibit remarkable diversity and include modulation of leukocyte activity and survival, cytokine and chemokine expression, maturation of antigen-presenting cells, antigen processing and presentation, B-cell selection and IgG affinity maturation, as well as regulation of IgG production. These functions are initiated upon specific interactions of the Fc domain with the various types of FcγRs-a process that is largely determined by the structural heterogeneity of the IgG Fc domain. Modulation of the Fc-associated glycan structure and composition along with differences in the primary amino acid sequence among the IgG subclasses represent the two main diversification mechanisms of the Fc domain that generate a spectrum of Fc domain phenotypes with distinct affinity for the various FcγR types and differential capacity to activate immunomodulatory pathways.
Collapse
Affiliation(s)
- Stylianos Bournazos
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
22
|
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
|
23
|
Abstract
Autoimmune diseases are characterized by adaptive immune responses against self-antigens, including humoral responses resulting in the production of autoantibodies. Autoantibodies generate inflammation by activating complement and engaging Fcγ receptors (FcγRs). The inhibitory receptor FcγRIIB plays a central role in regulating the generation of autoantibodies and their effector functions, which include activation of innate immune cells and the cellular arm of the adaptive immune system, via effects on antigen presentation to CD4 T cells. Polymorphisms in FcγRIIB have been associated with susceptibility to autoimmunity but protection against infections in humans and mice. In the last few years, new mechanisms by which FcγRIIB controls the adaptive immune response have been described. Notably, FcγRIIB has been shown to regulate germinal center B cells and dendritic cell migration, with potential impact on the development of autoimmune diseases. Recent work has also highlighted the implication of FcγRIIB on the regulation of the innate immune system, via inhibition of Toll-like receptor- and complement receptor-mediated activation. This review will provide an update on the role of FcγRIIB in adaptive immune responses in autoimmunity, and then focus on their emerging function in innate immunity.
Collapse
Affiliation(s)
- Marion Espéli
- Inserm UMR_S996, LabEx LERMIT, Université Paris-Sud, Paris, France
| | - Kenneth G C Smith
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Menna R Clatworthy
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| |
Collapse
|
24
|
Julián MT, Alonso N, Colobran R, Sánchez A, Miñarro A, Pujol-Autonell I, Carrascal J, Rodríguez-Fernández S, Ampudia RM, Vives-Pi M, Puig-Domingo M. CD26/DPPIV inhibition alters the expression of immune response-related genes in the thymi of NOD mice. Mol Cell Endocrinol 2016; 426:101-12. [PMID: 26911933 DOI: 10.1016/j.mce.2016.02.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 02/15/2016] [Accepted: 02/15/2016] [Indexed: 12/22/2022]
Abstract
The transmembrane glycoprotein CD26 or dipeptidyl peptidase IV (DPPIV) is a multifunctional protein. In immune system, CD26 plays a role in T-cell function and is also involved in thymic maturation and emigration patterns. In preclinical studies, treatment with DPPIV inhibitors reduces insulitis and delays or even reverses the new -onset of type 1 diabetes (T1D) in non-obese diabetic (NOD) mice. However, the specific mechanisms involved in these effects remain unknown. The aim of the present study was to investigate how DPPIV inhibition modifies the expression of genes in the thymus of NOD mice by microarray analysis. Changes in the gene expression of β-cell autoantigens and Aire in thymic epithelial cells (TECs) were also evaluated by using qRT-PCR. A DPPIV inhibitor, MK626, was orally administered in the diet for 4 and 6 weeks starting at 6-8 weeks of age. Thymic glands from treated and control mice were obtained for each study checkpoint. Thymus transcriptome analysis revealed that 58 genes were significantly over-expressed in MK626-treated mice after 6 weeks of treatment. Changes in gene expression in the thymus were confined mainly to the immune system, including innate immunity, chemotaxis, antigen presentation and immunoregulation. Most of the genes are implicated in central tolerance mechanisms through several pathways. No differences were observed in the expression of Aire and β-cell autoantigens in TECs. In the current study, we demonstrate that treatment with the DPPIV inhibitor MK626 in NOD mice alters the expression of the immune response-related genes in the thymus, especially those related to immunological central tolerance, and may contribute to the prevention of T1D.
Collapse
Affiliation(s)
- María Teresa Julián
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Health Sciences Research Institute and Hospital, 08916, Badalona, Spain; Department of Medicine, Autonomous University of Barcelona, 08193, Barcelona, Spain
| | - Núria Alonso
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Health Sciences Research Institute and Hospital, 08916, Badalona, Spain; Department of Medicine, Autonomous University of Barcelona, 08193, Barcelona, Spain; CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Roger Colobran
- Immunology Division, Vall d'Hebron Research Institute (VHIR), Vall d'Hebron University Hospital, 08035, Barcelona, Spain
| | - Alex Sánchez
- Statistics Department, Faculty of Biology, University of Barcelona, 08028, Barcelona, Spain; Statistics and Bioinformatics Unit, Vall d'Hebron Research Institute (VHIR), 08035, Barcelona, Spain
| | - Antoni Miñarro
- Statistics Department, Faculty of Biology, University of Barcelona, 08028, Barcelona, Spain
| | - Irma Pujol-Autonell
- Immunology Department, Germans Trias i Pujol Health Sciences Research Institute, 08916, Badalona, Autonomous University of Barcelona, Spain
| | - Jorge Carrascal
- Immunology Department, Germans Trias i Pujol Health Sciences Research Institute, 08916, Badalona, Autonomous University of Barcelona, Spain
| | - Silvia Rodríguez-Fernández
- Immunology Department, Germans Trias i Pujol Health Sciences Research Institute, 08916, Badalona, Autonomous University of Barcelona, Spain
| | - Rosa María Ampudia
- Immunology Department, Germans Trias i Pujol Health Sciences Research Institute, 08916, Badalona, Autonomous University of Barcelona, Spain
| | - Marta Vives-Pi
- Immunology Department, Germans Trias i Pujol Health Sciences Research Institute, 08916, Badalona, Autonomous University of Barcelona, Spain; CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Manel Puig-Domingo
- Department of Endocrinology and Nutrition, Germans Trias i Pujol Health Sciences Research Institute and Hospital, 08916, Badalona, Spain; Department of Medicine, Autonomous University of Barcelona, 08193, Barcelona, Spain; CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain; CIBER of Rare Diseases (CIBERER), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain.
| |
Collapse
|
25
|
Sniping the scout: Targeting the key molecules in dendritic cell functions for treatment of autoimmune diseases. Pharmacol Res 2016; 107:27-41. [DOI: 10.1016/j.phrs.2016.02.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 02/23/2016] [Accepted: 02/23/2016] [Indexed: 02/07/2023]
|
26
|
Fc Receptors and Fc Receptor-Like Molecules within the Immunoreceptor Family. ENCYCLOPEDIA OF IMMUNOBIOLOGY 2016. [PMCID: PMC7152311 DOI: 10.1016/b978-0-12-374279-7.02017-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Receptors for the Fc portion of immunoglobulins (FcRs) account for most cell-mediated biological activities of antibodies. The majority of FcRs are encoded by a set of genes, clustered in the fcr locus, on chromosome 1 in humans and on chromosome 1 and 3 in mice. Eight (in humans) and six (in mice) new genes were found, intermixed with FcR genes in corresponding fcr loci, which encode FcR-like molecules (FcRLs). FcRs and FcRLs are genetically, phylogenetically, structurally, and functionally related. FcRs and FcRLs, however, markedly differ by their ligands, their tissue distribution, and, therefore, by the biological functions they control. A systematic comparison of their biological properties leads to the conclusion that FcRLs are not like FcRs. They altogether form a single family within the immunoreceptor family, whose members fulfill distinct but complementary roles in immunity by differentially controlling innate and adaptive responses.
Collapse
|
27
|
Abstract
IgG antibodies are actively produced in response to antigenic challenge or passively administered as an effective form of immunotherapy to confer immunity against foreign antigens. Their protective activity is mediated through their bifunctional nature: a variable Fab domain mediates antigen-binding specificity, whereas the constant Fc domain engages Fcγ receptors (FcγRs) expressed on the surface of leukocytes to mediate effector functions. While traditionally considered the invariant domain of an IgG molecule, the Fc domain displays remarkable structural heterogeneity determined primarily by differences in the amino acid sequence of the various IgG subclasses and by the composition of the complex, Fc-associated biantennary N-linked glycan. These structural determinants regulate the conformational flexibility of the IgG Fc domain and affect its capacity to interact with distinct types of FcγRs (type I or type II FcγRs). FcγR engagement activates diverse downstream immunomodulatory pathways with pleiotropic functional consequences including cytotoxicity and phagocytosis of IgG-coated targets, differentiation and activation of antigen presenting cells, modulation of T-cell activation, plasma cell survival, and regulation of antibody responses. These functions highlight the importance of FcγR-mediated pathways in the modulation of adaptive immune responses and suggest a central role for IgG-FcγR interactions during active and passive immunization.
Collapse
Affiliation(s)
- Stylianos Bournazos
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, 1230 York Ave, New York, NY 10065
| | - Jeffrey V. Ravetch
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, 1230 York Ave, New York, NY 10065
| |
Collapse
|
28
|
Regulatory dendritic cells in autoimmunity: A comprehensive review. J Autoimmun 2015; 63:1-12. [PMID: 26255250 DOI: 10.1016/j.jaut.2015.07.011] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 07/17/2015] [Accepted: 07/23/2015] [Indexed: 12/31/2022]
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells (APC) with significant phenotypic heterogeneity and functional plasticity. DCs play crucial roles in initiating effective adaptive immune responses for elimination of invading pathogens and also in inducing immune tolerance toward harmless components to maintain immune homeostasis. The regulatory capacity of DCs depends on their immature state and distinct subsets, yet not restricted to the immature state and one specialized subset. The tolerogenicity of DC is controlled by a complex network of environmental signals and cellular intrinsic mechanisms. Regulatory DCs play an important role in the maintenance of immunological tolerance via the induction of T cell unresponsiveness or apoptosis, and generation of regulatory T cells. DCs play essential roles in driving autoimmunity via promoting the activation of effector T cells such as T helper 1 and T helper 17 cells, and/or suppressing the generation of regulatory T cells. Besides, a breakdown of DCs-mediated tolerance due to abnormal environmental signals or breakdown of intrinsic regulatory mechanisms is closely linked with the pathogenesis of autoimmune diseases. Novel immunotherapy taking advantage of the tolerogenic potential of regulatory DCs is being developed for treatment of autoimmune diseases. In this review, we will describe the current understanding on the generation of regulatory DC and the role of regulatory DCs in promoting tolerogenic immune responses and suppressing autoimmune responses. The emerging roles of DCs dysfunction in the pathogenesis of autoimmune diseases and the potential application of regulatory DCs in the treatment of autoimmune diseases will also be discussed.
Collapse
|
29
|
Park SH, Chen WC, Durmus N, Bleck B, Reibman J, Riemekasten G, Grunig G. The Effects of Antigen-Specific IgG1 Antibody for the Pulmonary-Hypertension-Phenotype and B Cells for Inflammation in Mice Exposed to Antigen and Fine Particles from Air Pollution. PLoS One 2015; 10:e0129910. [PMID: 26079807 PMCID: PMC4469456 DOI: 10.1371/journal.pone.0129910] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 05/14/2015] [Indexed: 12/14/2022] Open
Abstract
Air pollution is known to exacerbate chronic inflammatory conditions of the lungs including pulmonary hypertension, cardiovascular diseases and autoimmune diseases. Directly pathogenic antibodies bind pro-inflammatory cell receptors and cause or exacerbate inflammation. In contrast, anti-inflammatory antibody isotypes (e.g. mouse immunoglobulin G1, IgG1) bind inhibitory cell receptors and can inhibit inflammation. Our previous studies showed that co-exposure to antigen and urban ambient particulate matter (PM2.5) induced severe pulmonary arterial thickening and increased right ventricular systolic pressures in mice via T-cell produced cytokines, Interleukin (IL)-13 and IL-17A. The aim of the current study was to understand how B cell and antibody responses integrate into this T cell cytokine network for the pulmonary hypertension phenotype. Special focus was on antigen-specific IgG1 that is the predominant antibody in the experimental response to antigen and urban ambient PM2.5. Wild type and B cell-deficient mice were primed with antigen and then challenged with antigen and urban particulate matter and injected with antibodies as appropriate. Our data surprisingly showed that B cells were necessary for the development of increased right ventricular pressures and molecular changes in the right heart in response to sensitization and intranasal challenge with antigen and PM2.5. Further, our studies showed that both, the increase in right ventricular systolic pressure and right ventricular molecular changes were restored by reconstituting the B cell KO mice with antigen specific IgG1. In addition, our studies identified a critical, non-redundant role of B cells for the IL-17A-directed inflammation in response to exposure with antigen and PM2.5, which was not corrected with antigen-specific IgG1. In contrast, IL-13-directed inflammatory markers, as well as severe pulmonary arterial remodeling induced by challenge with antigen and PM2.5 were similar in B cell-deficient and wild type mice. Our studies have identified B cells and antigen specific IgG1 as potential therapeutic targets for pulmonary hypertension associated with immune dysfunction and environmental exposures.
Collapse
Affiliation(s)
- Sung-Hyun Park
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, New York, United States of America
| | - Wen-Chi Chen
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, New York, United States of America
| | - Nedim Durmus
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, New York, United States of America
| | - Bertram Bleck
- Department of Medicine, Division of Pulmonary Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Joan Reibman
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, New York, United States of America
- Department of Medicine, Division of Pulmonary Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | | | - Gabriele Grunig
- Department of Environmental Medicine, New York University Langone Medical Center, Tuxedo, New York, United States of America
- Department of Medicine, Division of Pulmonary Medicine, New York University Langone Medical Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
30
|
Schinnerling K, Soto L, García-González P, Catalán D, Aguillón JC. Skewing dendritic cell differentiation towards a tolerogenic state for recovery of tolerance in rheumatoid arthritis. Autoimmun Rev 2015; 14:517-27. [PMID: 25633325 DOI: 10.1016/j.autrev.2015.01.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 01/20/2015] [Indexed: 12/14/2022]
Abstract
To date, the available options to treat autoimmune diseases such as rheumatoid arthritis (RA) include traditional corticoids and biological drugs, which are not exempt of adverse effects. The development of cellular therapies based on dendritic cells with tolerogenic functions (TolDCs) has opened a new possibility to efficiently eradicate symptoms and control the immune response in the field of autoimmunity. TolDCs are an attractive tool for antigen-specific immunotherapy to restore self-tolerance in RA and other autoimmune disorders. A promising strategy is to inject autologous self-antigen-loaded TolDCs, which are able to delete or reprogram autoreactive T cells. Different protocols for the generation of stable human TolDCs have been established and the therapeutic effect of TolDCs has been investigated in multiple rodent models of arthritis. Pilot studies in humans confirmed that TolDC application is safe, encouraging clinical trials using self-antigen-loaded TolDCs in RA patients. Although an abundance of molecular regulators of DC functions has been discovered in the last decade, no master regulator of tolerogenicity has been identified yet. Further research is required to define biomarkers or key regulators of tolerogenicity that might facilitate the induction and monitoring of TolDCs.
Collapse
Affiliation(s)
- Katina Schinnerling
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Lilian Soto
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Paulina García-González
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Diego Catalán
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
| | - Juan C Aguillón
- Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile; Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
| |
Collapse
|
31
|
FcγRIIB regulates T-cell autoreactivity, ANCA production, and neutrophil activation to suppress anti-myeloperoxidase glomerulonephritis. Kidney Int 2014; 86:1140-9. [DOI: 10.1038/ki.2014.189] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 03/18/2014] [Accepted: 04/03/2014] [Indexed: 01/09/2023]
|
32
|
Chan J, Mehta S, Bharrhan S, Chen Y, Achkar JM, Casadevall A, Flynn J. The role of B cells and humoral immunity in Mycobacterium tuberculosis infection. Semin Immunol 2014; 26:588-600. [PMID: 25458990 PMCID: PMC4314354 DOI: 10.1016/j.smim.2014.10.005] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 10/09/2014] [Accepted: 10/10/2014] [Indexed: 12/24/2022]
Abstract
Mycobacterium tuberculosis remains a major public health burden. It is generally thought that while B cell- and antibody-mediated immunity plays an important role in host defense against extracellular pathogens, the primary control of intracellular microbes derives from cellular immune mechanisms. Studies on the immune regulatory mechanisms during infection with M. tuberculosis, a facultative intracellular organism, has established the importance of cell-mediated immunity in host defense during tuberculous infection. Emerging evidence suggest a role for B cell and humoral immunity in the control of intracellular pathogens, including obligatory species, through interactions with the cell-mediated immune compartment. Recent studies have shown that B cells and antibodies can significantly impact on the development of immune responses to the tubercle bacillus. In this review, we present experimental evidence supporting the notion that the importance of humoral and cellular immunity in host defense may not be entirely determined by the niche of the pathogen. A comprehensive approach that examines both humoral and cellular immunity could lead to better understanding of the immune response to M. tuberculosis.
Collapse
Affiliation(s)
- John Chan
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Simren Mehta
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sushma Bharrhan
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yong Chen
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jacqueline M Achkar
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Arturo Casadevall
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - JoAnne Flynn
- Departments of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
33
|
Rabaglino MB, Keller-Wood M, Wood CE. Transcriptomics of the late gestation ovine fetal brain: modeling the co-expression of immune marker genes. BMC Genomics 2014; 15:1001. [PMID: 25409740 PMCID: PMC4253626 DOI: 10.1186/1471-2164-15-1001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 10/28/2014] [Indexed: 01/10/2023] Open
Abstract
Background Major changes in gene expression occur in the fetal brain to modulate the function of this organ postnatally. Thus, factors can alter the genomics of the fetal brain, predisposing to neurological disorders later in life. We hypothesized that the physiological dynamics of the immune system transcriptome of the fetal brain during the last stage of gestation will reveal patterns of immune function and development in the developing brain. In this study we applied weighted gene co-expression analysis of microarrays performed on ovine fetal brain samples, to model the changes in gene expression throughout the second half of gestation. Results Clusters of co-expressed genes that strongly increase in expression toward the first day of extra-uterine life are related to the hematopoietic lineage, while activation of immune pathways is induced after birth. Moreover, the pattern of gene expression suggests induction of tolerance mechanisms, probably necessary to protect highly produced proteins –such as myelin basic protein- from an autoimmune attack. Conclusions This study provides insight into the dramatic changes in gene expression that take place in the brain during the fetal life, especially during the last stage of gestation, and suggests that the immune system may have an important role in maturation of the fetal brain, which if disrupted or altered, could have negative consequences in postnatal life. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-1001) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maria B Rabaglino
- Departamento de Reproducción Animal, Fac, Agronomía y Veterinaria, Universidad Nacional de Río Cuarto, Córdoba, Argentina.
| | | | | |
Collapse
|
34
|
Furlan SN, Mandraju R, Brewer T, Roybal K, Troutman TD, Hu W, Palm NW, Unni A, Pasare C. Enhancement of anti-tumor CD8 immunity by IgG1-mediated targeting of Fc receptors. MAbs 2014; 6:108-18. [PMID: 24284965 DOI: 10.4161/mabs.27052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Dendritic cells (DCs) function as professional antigen presenting cells and are critical for linking innate immune responses to the induction of adaptive immunity. Many current cancer DC vaccine strategies rely on differentiating DCs, feeding them tumor antigens ex vivo, and infusing them into patients. Importantly, this strategy relies on prior knowledge of suitable “tumor-specific” antigens to prime an effective anti-tumor response. DCs express a variety of receptors specific for the Fc region of immunoglobulins, and antigen uptake via Fc receptors is highly efficient and facilitates antigen presentation to T cells. Therefore, we hypothesized that expression of the mouse IgG1 Fc region on the surface of tumors would enhance tumor cell uptake by DCs and other myeloid cells and promote the induction of anti-tumor T cell responses. To test this, we engineered a murine lymphoma cell line expressing surface IgG1 Fc and discovered that such tumor cells were taken up rapidly by DCs, leading to enhanced cross-presentation of tumor-derived antigen to CD8+ T cells. IgG1-Fc tumors failed to grow in vivo and prophylactic vaccination of mice with IgG1-Fc tumors resulted in rejection of unmanipulated tumor cells. Furthermore, IgG1-Fc tumor cells were able to slow the growth of an unmanipulated primary tumor when used as a therapeutic tumor vaccine. Our data demonstrate that engagement of Fc receptors by tumors expressing the Fc region of IgG1 is a viable strategy to induce efficient and protective anti-tumor CD8+ T cell responses without prior knowledge of tumor-specific antigens.
Collapse
|
35
|
Pincetic A, Bournazos S, DiLillo DJ, Maamary J, Wang TT, Dahan R, Fiebiger BM, Ravetch JV. Type I and type II Fc receptors regulate innate and adaptive immunity. Nat Immunol 2014; 15:707-16. [PMID: 25045879 DOI: 10.1038/ni.2939] [Citation(s) in RCA: 386] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 06/09/2014] [Indexed: 12/15/2022]
Abstract
Antibodies produced in response to a foreign antigen are characterized by polyclonality, not only in the diverse epitopes to which their variable domains bind but also in the various effector molecules to which their constant regions (Fc domains) engage. Thus, the antibody's Fc domain mediates diverse effector activities by engaging two distinct classes of Fc receptors (type I and type II) on the basis of the two dominant conformational states that the Fc domain may adopt. These conformational states are regulated by the differences among antibody subclasses in their amino acid sequence and by the complex, biantennary Fc-associated N-linked glycan. Here we discuss the diverse downstream proinflammatory, anti-inflammatory and immunomodulatory consequences of the engagement of type I and type II Fc receptors in the context of infectious, autoimmune, and neoplastic disorders.
Collapse
Affiliation(s)
- Andrew Pincetic
- 1] The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA. [2]
| | - Stylianos Bournazos
- 1] The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA. [2]
| | - David J DiLillo
- 1] The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA. [2]
| | - Jad Maamary
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA
| | - Taia T Wang
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA
| | - Rony Dahan
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA
| | | | - Jeffrey V Ravetch
- The Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA
| |
Collapse
|
36
|
Platzer B, Stout M, Fiebiger E. Antigen cross-presentation of immune complexes. Front Immunol 2014; 5:140. [PMID: 24744762 PMCID: PMC3978348 DOI: 10.3389/fimmu.2014.00140] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 03/19/2014] [Indexed: 12/23/2022] Open
Abstract
The ability of dendritic cells (DCs) to cross-present tumor antigens has long been a focus of interest to physicians, as well as basic scientists, that aim to establish efficient cell-based cancer immune therapy. A prerequisite for exploiting this pathway for therapeutic purposes is a better understanding of the mechanisms that underlie the induction of tumor-specific cytotoxic T-lymphocyte (CTL) responses when initiated by DCs via cross-presentation. The ability of humans DC to perform cross-presentation is of utmost interest, as this cell type is a main target for cell-based immunotherapy in humans. The outcome of a cross-presentation event is guided by the nature of the antigen, the form of antigen uptake, and the subpopulation of DCs that performs presentation. Generally, CD8α+ DCs are considered to be the most potent cross-presenting DCs. This paradigm, however, only applies to soluble antigens. During adaptive immune responses, immune complexes form when antibodies interact with their specific epitopes on soluble antigens. Immunoglobulin G (IgG) immune complexes target Fc-gamma receptors on DCs to shuttle exogenous antigens efficiently into the cross-presentation pathway. This receptor-mediated cross-presentation pathway is a well-described route for the induction of strong CD8+ T cell responses. IgG-mediated cross-presentation is intriguing because it permits the CD8− DCs, which are commonly considered to be weak cross-presenters, to efficiently cross-present. Engaging multiple DC subtypes for cross-presentation might be a superior strategy to boost CTL responses in vivo. We here summarize our current understanding of how DCs use IgG-complexed antigens for the efficient induction of CTL responses. Because of its importance for human cell therapy, we also review the recent advances in the characterization of cross-presentation properties of human DC subsets.
Collapse
Affiliation(s)
- Barbara Platzer
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Boston Children's Hospital, Harvard Medical School , Boston, MA , USA
| | - Madeleine Stout
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Boston Children's Hospital, Harvard Medical School , Boston, MA , USA
| | - Edda Fiebiger
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Boston Children's Hospital, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
37
|
Guilliams M, Bruhns P, Saeys Y, Hammad H, Lambrecht BN. The function of Fcγ receptors in dendritic cells and macrophages. Nat Rev Immunol 2014; 14:94-108. [PMID: 24445665 DOI: 10.1038/nri3582] [Citation(s) in RCA: 492] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Dendritic cells (DCs) and macrophages use various receptors to recognize foreign antigens and to receive feedback control from adaptive immune cells. Although it was long believed that all immunoglobulin Fc receptors are universally expressed by phagocytes, recent findings indicate that only monocyte-derived DCs and macrophages express high levels of activating Fc receptors for IgG (FcγRs), whereas conventional and plasmacytoid DCs express the inhibitory FcγR. In this Review, we discuss how the uptake, processing and presentation of antigens by DCs and macrophages is influenced by FcγR recognition of immunoglobulins and immune complexes in the steady state and during inflammation.
Collapse
Affiliation(s)
- Martin Guilliams
- 1] Laboratory of Immunoregulation, VIB Inflammation Research Center, 9052 Ghent, Belgium. [2] Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Pierre Bruhns
- 1] Institut Pasteur, Département d'Immunologie, Laboratoire Anticorps en Thérapie et Pathologie, 75015 Paris, France. [2] Institut National de la Santé et de la Recherche Médicale, U760, 75015 Paris, France
| | - Yvan Saeys
- 1] Laboratory of Immunoregulation, VIB Inflammation Research Center, 9052 Ghent, Belgium. [2] Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Hamida Hammad
- 1] Laboratory of Immunoregulation, VIB Inflammation Research Center, 9052 Ghent, Belgium. [2] Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Bart N Lambrecht
- 1] Laboratory of Immunoregulation, VIB Inflammation Research Center, 9052 Ghent, Belgium. [2] Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium. [3] Department of Pulmonary Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
| |
Collapse
|
38
|
Lehmann B, Schwab I, Böhm S, Lux A, Biburger M, Nimmerjahn F. FcγRIIB: a modulator of cell activation and humoral tolerance. Expert Rev Clin Immunol 2014; 8:243-54. [DOI: 10.1586/eci.12.5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
39
|
White AL, Beers SA, Cragg MS. FcγRIIB as a key determinant of agonistic antibody efficacy. Curr Top Microbiol Immunol 2014; 382:355-72. [PMID: 25116108 DOI: 10.1007/978-3-319-07911-0_16] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Fc gamma Receptor (FcγR) IIB (CD32B) is an immunoreceptor tyrosine inhibitory motif (ITIM)-bearing Fc receptor that is involved in abrogating the signalling and function delivered from other receptors; archetypally those arising from other, activatory, FcγR and from the B cell receptor (BCR) for antigen. In the context of immunotherapy, it has convincingly been shown to limit a variety of clinically important therapeutic monoclonal antibodies (mAb) such as rituximab and trastuzumab in preclinical models. However, recent exploration of so-called immunomodulatory mAb, for example agonist mAb directed against various members of the TNFR super-family, has cast new light on the ability of FcγRIIB to regulate immune responses and immunotherapy. These data, accumulated by several independent groups, have shown the seemingly paradoxical ability of FcγRIIB to augment or even be absolutely required for the activity of this class of mAb. In this review we highlight the key role of FcγRIIB in regulating agonistic mAb, detail the likely mechanism of action and propose new ways in which this information may be exploited therapeutically.
Collapse
Affiliation(s)
- Ann L White
- Cancer Sciences Unit, Antibody and Vaccine Group (MP88), Faculty of Medicine, Southampton University, Tremona Road, Southampton, SO16 6YD, UK
| | | | | |
Collapse
|
40
|
Abstract
Antibodies are major molecular effectors of adaptive immune responses. Most, if not all, biological activities of antibodies, however, depend on the functional properties of cells that express receptors for the Fc portion of antibodies (FcR). Most FcR are activating receptors; some are inhibitory. When engaged by antibodies and antigen, the various FcR expressed by a given cell trigger a mixture of positive and negative signals whose integration determines cellular responses. Responses of cell populations can be either protective or pathogenic. As a consequence, FcR are potential target/tools in a variety of diseases including infection, allergy, autoimmune diseases, and cancer.
Collapse
|
41
|
Mimoto F, Katada H, Kadono S, Igawa T, Kuramochi T, Muraoka M, Wada Y, Haraya K, Miyazaki T, Hattori K. Engineered antibody Fc variant with selectively enhanced FcγRIIb binding over both FcγRIIa(R131) and FcγRIIa(H131). Protein Eng Des Sel 2013; 26:589-98. [PMID: 23744091 PMCID: PMC3785249 DOI: 10.1093/protein/gzt022] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 04/24/2013] [Accepted: 05/08/2013] [Indexed: 12/02/2022] Open
Abstract
Engaging inhibitory FcγRIIb by Fc region has been recently reported to be an attractive approach for improving the efficacy of antibody therapeutics. However, the previously reported S267E/L328F variant with enhanced binding affinity to FcγRIIb, also enhances binding affinity to FcγRIIa(R131) allotype to a similar degree because FcγRIIb and FcγRIIa(R131) are structurally similar. In this study, we applied comprehensive mutagenesis and structure-guided design based on the crystal structure of the Fc/FcγRIIb complex to identify a novel Fc variant with selectively enhanced FcγRIIb binding over both FcγRIIa(R131) and FcγRIIa(H131). This novel variant has more than 200-fold stronger binding affinity to FcγRIIb than wild-type IgG1, while binding affinity to FcγRIIa(R131) and FcγRIIa(H131) is comparable with or lower than wild-type IgG1. This selectivity was achieved by conformational change of the C(H)2 domain by mutating Pro to Asp at position 238. Fc variant with increased binding to both FcγRIIb and FcγRIIa induced platelet aggregation and activation in an immune complex form in vitro while our novel variant did not. When applied to agonistic anti-CD137 IgG1 antibody, our variant greatly enhanced the agonistic activity. Thus, the selective enhancement of FcγRIIb binding achieved by our Fc variant provides a novel tool for improving the efficacy of antibody therapeutics.
Collapse
Affiliation(s)
| | | | | | - T. Igawa
- Research Division, Chugai Pharmaceutical Co., Ltd., Gotemba, Shizuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
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
|
43
|
Kozakiewicz L, Phuah J, Flynn J, Chan J. The role of B cells and humoral immunity in Mycobacterium tuberculosis infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 783:225-50. [PMID: 23468112 DOI: 10.1007/978-1-4614-6111-1_12] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tuberculosis (TB) remains a serious threat to public health, causing 2 million deaths annually world-wide. The control of TB has been hindered by the requirement of long duration of treatment involving multiple chemotherapeutic agents, the increased susceptibility to Mycobacterium tuberculosis infection in the HIV-infected population, and the development of multi-drug resistant and extensively resistant strains of tubercle bacilli. An efficacious and cost-efficient way to control TB is the development of effective anti-TB vaccines. This measure requires thorough understanding of the immune response to M. tuberculosis. While the role of cell-mediated immunity in the development of protective immune response to the tubercle bacillus has been well established, the role of B cells in this process is not clearly understood. Emerging evidence suggests that B cells and humoral immunity can modulate the immune response to various intracellular pathogens, including M. tuberculosis. These lymphocytes form conspicuous aggregates in the lungs of tuberculous humans, non-human primates, and mice, which display features of germinal center B cells. In murine TB, it has been shown that B cells can regulate the level of granulomatous reaction, cytokine production, and the T cell response. This chapter discusses the potential mechanisms by which specific functions of B cells and humoral immunity can shape the immune response to intracellular pathogens in general, and to M. tuberculosis in particular. Knowledge of the B cell-mediated immune response to M. tuberculosis may lead to the design of novel strategies, including the development of effective vaccines, to better control TB.
Collapse
Affiliation(s)
- Lee Kozakiewicz
- Department of Medicine and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | | | |
Collapse
|
44
|
Overcoming Resistance to Therapeutic Antibodies by Targeting Fc Receptors. RESISTANCE TO TARGETED ANTI-CANCER THERAPEUTICS 2013. [DOI: 10.1007/978-1-4614-7654-2_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
45
|
Callaghan CJ, Win TS, Motallebzadeh R, Conlon TM, Chhabra M, Harper I, Sivaganesh S, Bolton EM, Bradley JA, Brownlie RJ, Smith KGC, Pettigrew GJ. Regulation of allograft survival by inhibitory FcγRIIb signaling. THE JOURNAL OF IMMUNOLOGY 2012; 189:5694-702. [PMID: 23150718 DOI: 10.4049/jimmunol.1202084] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Fcγ receptors (FcγR) provide important immunoregulation. Targeting inhibitory FcγRIIb may therefore prolong allograft survival, but its role in transplantation has not been addressed. FcγRIIb signaling was examined in murine models of acute or chronic cardiac allograft rejection by transplanting recipients that either lacked FcγRIIb expression (FcγRIIb(-/-)) or overexpressed FcγRIIb on B cells (B cell transgenic [BTG]). Acute heart allograft rejection occurred at the same tempo in FcγRIIb(-/-) C57BL/6 (B6) recipients as wild type recipients, with similar IgG alloantibody responses. In contrast, chronic rejection of MHC class II-mismatched bm12 cardiac allografts was accelerated in FcγRIIb(-/-) mice, with development of more severe transplant arteriopathy and markedly augmented effector autoantibody production. Autoantibody production was inhibited and rejection was delayed in BTG recipients. Similarly, whereas MHC class I-mismatched B6.K(d) hearts survived indefinitely and remained disease free in B6 mice, much stronger alloantibody responses and progressive graft arteriopathy developed in FcγRIIb(-/-) recipients. Notably, FcγRIIb-mediated inhibition of B6.K(d) heart graft rejection was abrogated by increasing T cell help through transfer of additional H2.K(d)-specific CD4 T cells. Thus, inhibitory FcγRIIb signaling regulates chronic but not acute rejection, most likely because the supra-optimal helper CD4 T cell response in acute rejection overcomes FcγRIIb-mediated inhibition of the effector B cell population. Immunomodulation of FcγRIIb in clinical transplantation may hold potential for inhibiting progression of transplant arteriopathy and prolonging transplant survival.
Collapse
Affiliation(s)
- Chris J Callaghan
- Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
van Montfoort N, ’t Hoen PAC, Mangsbo SM, Camps MGM, Boross P, Melief CJM, Ossendorp F, Verbeek JS. Fcγ Receptor IIb Strongly Regulates Fcγ Receptor-Facilitated T Cell Activation by Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2012; 189:92-101. [DOI: 10.4049/jimmunol.1103703] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
47
|
Fu J, Zhang A, Ju X. Tolerogenic dendritic cells as a target for the therapy of immune thrombocytopenia. Clin Appl Thromb Hemost 2012; 18:469-75. [PMID: 22387587 DOI: 10.1177/1076029612438612] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disease in which platelets are destroyed by special antiplatelet autoantibodies produced by B cells. Dendritic cells (DCs) are professional antigen-presenting cells involved in humoral immunity and cellular immunity and among them DCs that induce autoimmune tolerance are called tolerogenic DCs (tDCs). As a promising immunotherapeutic strategy for ITP, tDCs have received increasing attention. In this review, we describe the significant role of DCs in regulating autoimmune balances, introduce the manipulation strategies to generate tDCs, summarize recent progress on the experimental application of tDCs for ITP therapy, and finally discuss the perspectives of tolerogenic vaccination for ITP treatment in the clinic.
Collapse
Affiliation(s)
- Jinqiu Fu
- Shandong University, Shandong, China
| | | | | |
Collapse
|
48
|
Nordstrom JL, Gorlatov S, Zhang W, Yang Y, Huang L, Burke S, Li H, Ciccarone V, Zhang T, Stavenhagen J, Koenig S, Stewart SJ, Moore PA, Johnson S, Bonvini E. Anti-tumor activity and toxicokinetics analysis of MGAH22, an anti-HER2 monoclonal antibody with enhanced Fcγ receptor binding properties. Breast Cancer Res 2011; 13:R123. [PMID: 22129105 PMCID: PMC3326565 DOI: 10.1186/bcr3069] [Citation(s) in RCA: 191] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 07/20/2011] [Accepted: 11/30/2011] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Response to trastuzumab in metastatic breast cancer correlates with expression of the high binding variant (158V) of the activating Fcγ receptor IIIA (CD16A). We engineered MGAH22, a chimeric anti-HER2 monoclonal antibody with specificity and affinity similar to trastuzumab, with an Fc domain engineered for increased binding to both alleles of human CD16A. METHODS MGAH22 was compared to an identical anti-HER2 mAb except for a wild type Fc domain. Antibody-dependent cell cytotoxicity (ADCC) assays were performed with HER2-expressing cancer cells as targets and human PBMC or purified NK cells as effectors. Xenograft studies were conducted in mice with wild type murine FcγRs; in mice lacking murine CD16; or in mice lacking murine CD16 but transgenic for human CD16A-158F, the low-binding variant. The latter model reproduces the differential binding between wild type and the Fc-optimized mAb for human CD16A. The JIMT-1 human breast tumor line, derived from a patient that progressed on trastuzumab therapy, was used in these studies. Single and repeat dose toxicology studies with MGAH22 administered intravenously at high dose were conducted in cynomolgus monkeys. RESULTS The optimized Fc domain confers enhanced ADCC against all HER2-positive tumor cells tested, including cells resistant to trastuzumab's anti-proliferative activity or expressing low HER2 levels. The greatest improvement occurs with effector cells isolated from donors homozygous or heterozygous for CD16A-158F, the low-binding allele. MGAH22 demonstrates increased activity against HER2-expressing tumors in mice transgenic for human CD16A-158F. In single and repeat-dose toxicology studies in cynomolgus monkeys, a species with a HER2 expression pattern comparable to that in humans and Fcγ receptors that exhibit enhanced binding to the optimized Fc domain, MGAH22 was well tolerated at all doses tested (15-150 mg/kg) and exhibited pharmacokinetic parameters similar to that of other anti-HER2 antibodies. Induction of cytokine release by MGAH22 in vivo or in vitro was similar to that induced by the corresponding wild type mAb or trastuzumab. CONCLUSIONS The data support the clinical development of MGAH22, which may have utility in patients with low HER2 expressing tumors or carrying the CD16A low-binding allele.
Collapse
|
49
|
Abstract
One of the most fundamental problems in immunology is the seemingly schizophrenic ability of the immune system to launch robust immunity against pathogens, while acquiring and maintaining a state of tolerance to the body's own tissues and the trillions of commensal microorganisms and food antigens that confront it every day. A fundamental role for the innate immune system, particularly dendritic cells (DCs), in orchestrating immunological tolerance has been appreciated, but emerging studies have highlighted the nature of the innate receptors and the signaling pathways that program DCs to a tolerogenic state. Furthermore, several studies have emphasized the major role played by cellular interactions and the microenvironment in programming tolerogenic DCs. Here, we review these studies and suggest that the innate control of tolerogenic responses can be viewed as different hierarchies of organization, in which DCs, their innate receptors and signaling networks, and their interactions with other cells and local microenvironments represent different levels of the hierarchy.
Collapse
Affiliation(s)
- Santhakumar Manicassamy
- Emory Vaccine Center, Yerkes National Primate Research Center, Department of Pathology, Emory University School of Medicine, Atlanta, GA 30329, USA
| | | |
Collapse
|
50
|
Haasken S, Auger JL, Binstadt BA. Absence of β2 integrins impairs regulatory T cells and exacerbates CD4+ T cell-dependent autoimmune carditis. THE JOURNAL OF IMMUNOLOGY 2011; 187:2702-10. [PMID: 21795599 DOI: 10.4049/jimmunol.1000967] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The immunopathogenic mechanisms mediating inflammation in multiorgan autoimmune diseases may vary between the different target tissues. We used the K/BxN TCR transgenic mouse model to investigate the contribution of CD4(+) T cells and β(2) integrins in the pathogenesis of autoimmune arthritis and endocarditis. Depletion of CD4(+) T cells following the onset of arthritis specifically prevented the development of cardiac valve inflammation. Genetic absence of β(2) integrins had no effect on the severity of arthritis and unexpectedly increased the extent of cardiovascular pathology. The exaggerated cardiac phenotype of the β(2) integrin-deficient K/BxN mice was accompanied by immune hyperactivation and was linked to a defect in regulatory T cells. These findings are consistent with a model in which the development of arthritis in K/BxN mice relies primarily on autoantibodies, whereas endocarditis depends on an additional contribution of effector T cells. Furthermore, strategies targeting β(2) integrins for the treatment of systemic autoimmune conditions need to consider not only the role of these molecules in leukocyte recruitment to sites of inflammation, but also their impact on the regulation of immunological tolerance.
Collapse
Affiliation(s)
- Stefanie Haasken
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA
| | | | | |
Collapse
|